You are on page 1of 16

Molecular Microbiology (2011) 81(6), 1577–1592 䊏 doi:10.1111/j.1365-2958.2011.07792.

x
First published online 23 August 2011

The Mycobacterium tuberculosis Ag85A is a novel


diacylglycerol acyltransferase involved in lipid
body formation mmi_7792 1577..1592

Ayssar A. Elamin,1 Matthias Stehr,1 Ralf Spallek,3 role in the formation of lipid storage bodies and thus
Manfred Rohde2 and Mahavir Singh1,3* also in the establishment and maintenance of a per-
Departments of 1Gene Regulation and Differentiation sistent tuberculosis infection.
and 2Medical Microbiology, Helmholtz Centre for
Infection Research, 38124 Braunschweig, Germany.
3 Introduction
LIONEX Diagnostics and Therapeutics GmbH, D-38126
Braunschweig, Germany. More than a third of the world’s population is infected with
Mycobacterium tuberculosis, the bacterium that causes
tuberculosis (TB). M. tuberculosis is the leading cause of
Summary death from a single bacterial species (Coker, 2004). TB
Mycobacterium tuberculosis accumulates large disease kills approximately 1.7 million people worldwide
amounts of triacylglycerol (TAG) which acts as annually (Russell et al., 2010). The emergence of new
storage compounds for energy and carbon. The strains of M. tuberculosis which have become resistant to
mycobacterial triacylglycerols stored in the form of standard antibiotic therapy has given urgency to the
intracellular lipid droplets are essential for long-term search for a better vaccine and new drugs against this
survival of M. tuberculosis during a dormant state. We pathogen (Bass et al., 1994; de Jong et al., 2004).
report here that when the M. tuberculosis myco- Mycobacterium tuberculosis shows a remarkable prop-
lytransferase Ag85A is overexpressed in Mycobacte- erty of existing in different states of invasion (infection),
rium smegmatis mc2155, cell morphology was colonization and persistence. It also has outstanding
changed and the cells became grossly enlarged. A mechanisms to escape from elimination and immune or
massive formation of lipid bodies and a change in lipid therapeutic eradication (Casadevall and Pirofski, 2000).
pattern was observed simultaneously. We suspected The major obstacle for host defence mechanisms (Jarlier
a possible role of Ag85A in the acyl lipid metabolism and Nikaido, 1994) and therapeutic intervention is the
and discovered that the enzyme possesses acyl- unusual robust cell wall of mycobacterium. The cell wall
CoA:diacylglycerol acyltransferase (DGAT) activity in structure of M. tuberculosis is unique among prokaryotes,
addition to its well-known function as mycolyltrans- and it is a major determinant of virulence for the bacterium
ferase. Ag85A mediates the transesterification of dia- (Belisle et al., 1997; Daffe and Draper, 1998). The inner
cylglycerol using long-chain acyl-CoA as acyl donors. peptidoglycan sacculus of the mycobacterial cell enve-
The Km and Kcat values for palmitoleoyl-coenzyme A lope is wrapped by a polysaccharide layer of mycolyl-
were 390 mM and 55.54 min-1 respectively. A docking arabinogalactan. The outer envelope consists of trehalose
model suggests that palmitoleoyl-coenzyme A and 6,6′-dimycolate (TDM; cord factor) where the mycolic
1,2-dipalmitin occupy the same active site as treha- acids of TDM interact with the mycolyl-residues from the
lose 6,6⬘-dimycolate and trehalose 6⬘-monomycolate. layer beneath. The mycolic acid-containing layers have
The site-directed Ser126Ala mutation of the active site width of ~10 nm and limit the penetration of hydrophilic
proved that this residue is involved in the catalytic substances, and water-soluble antibiotics, whereas the
activity of this enzyme. Although not proven conclu- inner saccharide layer inhibits the penetration of such
sively for dormant stage of M. tuberculosis, our novel substances. Due to the extremely low permeability of the
finding about the synthesis of TAGs by Ag85A cell wall mycobacteria show a high degree of intrinsic
strongly suggests that Ag85A may play a significant resistance to most antibiotics and chemotherapeutic
agents (Ducati et al., 2006). Thus the biosynthesis of
mycolic acids and TDM have become a major target of
Accepted 27 July, 2011. *For correspondence. E-mail msi@
helmholtz-hzi.de; Tel. (+49) 531 6181 5320/5309; Fax (+49) 531 drug research (Brennan and Nikaido, 1995). TDM is
6181 5399. exclusively synthesized by the enzymes of the antigen 85
© 2011 Blackwell Publishing Ltd
1578 A. A. Elamin et al. 䊏

(Ag85) complex Ag85A, B and C, which are acyltrans- Results


ferases and convert TMM (trehalose 6′-monomycolate)
to TDM (Belisle et al., 1997). Antigen 85C synthesizes Overexpression of Ag85A changes cell shape, size and
around 50% of the cell wall linked mycolic acid (Jackson results in formation of giant cells
et al., 1999) and antigen 85A has been shown to be crucial The recombinant M. smegmatis-Rv3804c showed rapid
for the growth in nutrient-poor media and macrophage-like overexpression of Ag85A from the mycobacterial heat
cell line (Armitige et al., 2000). shock promoter Phsp60 even before induction (Fig. 1) as
Therapeutic eradication of TB is affected by the ability of observed previously (Stover et al., 1991). After 18 h no
the bacterium to survive up to decades in a dormant state, further increase of expression was observed (Fig. 1I) and
primarily in hypoxic granulomas in the lung (Murphy and the recombinant M. smegmatis-Rv3804c showed pro-
Brown, 2007). Unfortunately, very little is known about the minent changes in cell size, morphology and surface
biology of the dormant phase and no specific persister structure. The increase in cell length correlated to the
genes have been identified yet (Cardona, 2007; Lewis, extended expression of Ag85A. A majority of the wild-type
2007; Rao and Li, 2009). This lack of well-defined targets M. smegmatis mc2155 cells were ~2 mm in length
specific to the dormancy phase has been a major obstacle (Fig. 1A) whereas M. smegmatis-Rv3804c cells increased
for the identification and development of novel leads in length up to 4–5 mm (Fig. 1B and C). These results
(Murphy and Brown, 2007). One important factor for long- indicated that expression of Ag85A interfered with cell
term survival of the bacillus seems to be the ability to store division and led to filamentation. Some of the elongated
and metabolize fatty acids, which are thought to be the cells showed buds and bulb-like structures (Fig. 1D and
major energy source in the dormant phase (Daniel et al., E). M. smegmatis-Rv3804c cells cultured in liquid media
2004). Genes that encode enzymes required to metabolize (7H9) containing Tween-80 showed significant clumping
fatty acids as the major carbon source, such as isocitrate and aggregation specifically in the late stationary growth
lyase, were found to be essential for persistence (McKin- phase (Fig. 1–2 and H), while the control M. smegmatis
ney et al., 2000; Meena and Rajni, 2010). Actinomycetes mc2155 did not show any tendency to clump (Fig. 1F1 and
synthesize and accumulate large amounts of triacylglyc- G). To investigate the possibility that the observed aggre-
erol (TAG) which act as storage compounds for energy and gation was not due to the presence of kanamycin, the
carbon (Olukoshi and Packter, 1994; Alvarez et al., 2000; M. smegmatis-pMV261 cells containing the empty vector
Alvarez and Steinbüchel, 2002). Mycobacteria store TAGs were cultured in the same condition. No aggregation phe-
in the form of intracellular lipid droplets during hypoxia- notypes or any changes in cell size were observed (data
induced non-replicating persistence (Garton et al., 2002; not shown).
Daniel et al., 2004). These bacteria are phenotypically Immunoelectron microscopy revealed that Ag85A is
drug-resistant and probably the cause for prolonged TB mostly localized in the inner side of cell membrane or cell
treatment required for therapy (Low et al., 2010). Lipid wall and at the surface of TAG inclusions (Fig. 2) as has
body positive acid fast bacilli have been suggested to be been found earlier in mycobacteria residing in phago-
a biomarker for non-replicating M. tuberculosis cells in somes (Beatty and Russell, 2000). Gold labelling of
sputum (Garton et al., 2008; Low et al., 2010). Out of 15 Ag85A showed that the enzyme is also present in the
putative TAG synthases (Tgs) (diacylglycerol acyltrans- cytoplasm.
ferase) genes known, only for tgs1 the involvement in
TAG accumulation has been proven experimentally
Overexpression of Ag85A induces lipid body formation
(Kalscheuer and Steinbüchel, 2003; Daniel et al., 2004;
and thickening of the cell wall in recombinant
Sirakova et al., 2006).
M. smegmatis
During our overexpression studies with the M. tubercu-
losis Ag85A in M. smegmatis mc2155 we observed a Mycobacterium smegmatis mc2155 and recombinant
massive formation of lipid bodies and a change of the lipid M. smegmatis-Rv3804c were cultured under similar
pattern, showing the involvement of Ag85A in TAG synthe- conditions as described in Experimental procedures
sis. Prompted by this finding we carried out detailed enzy- and samples were collected after induction. Transmis-
matic characterization of Ag85A and report for the first sion electron microscopic (TEM) on ultrathin sections of
time that M. tuberculosis Ag85A possess, in addition to its the cells revealed a significant change only in the in
mycolyltransferase activity, also diacylglycerol acyltrans- morphology in M. smegmatis-Rv3804c cells and concur-
ferase (DGAT) activity. This finding strongly suggests that rently the appearance of neutral lipid vesicles (Fig. 3A
Ag85A is a TAG synthase and might be a key player in both and B). Sections of M. smegmatis-Rv3804c showed an
cell wall stability and the biosynthesis of storage com- abundance of electron-transparent vesicles within the
pounds for the survival of M. tuberculosis in the dormant cytoplasm that had no internal structure. These discrete
state. structures appeared to be bordered by a thin membrane,

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1579

Fig. 1. Overexpression of Mycobacterium tuberculosis Ag85A altered the morphology of M. smegmatis cells.
A–H. FESEM analysis revealed enlargement and branching of M. smegmatis-Rv3804c (B and C) with changing of surface shape, also after
induction the cell exhibits tubular like structures (D and E). Wild-type M. smegmatis mc2155 was used as control (A and G). (F) M. smegmatis
mc2155 [1] M. smegmatis-Rv3804c [2] were cultured in 7H9 liquid media with ADC, 0.2% glycerol and 0.05% Tween 80 until an OD600 = 1.8
then the aggregation was tested by allowing the cells to settle for 15 min. The aggregation was clearly visible in FESEM images of
M. smegmatis-Rv3804c (H) while M. smegmatis mc2155 does not show any clumping (G).
I. Kinetics of Ag85A overexpression associated with induced changes in cell morphology by using SDS/PAGE on 12% (w/v) polyacrylamide
gels stained with Coomassie Brilliant Blue R-250. Molecular mass marker proteins were from Fermentas (L). Samples were collected from the
main culture at 12 (lane 2), 14 (lane 3), 16 (lane 4) and 18 h (lane 5) and also after 2 h induction (lane 6) at 45°C with 0.002% H2O2, wild-type
M. smegmatis mc2155 (lane 1) used as control.

were variable in sizes and occupied a substantial portion visible oleogenous layer as observed by Alvarez and
of the total cell volume (approximately 30–70%). Steinbüchel (2002). The PBs enlarged and dissociated
After induction, all intermediate stages of matured lipid into the cytoplasm forming RPBs. These RPBs enlarged
bodies (MLBs), lipid prebodies (PBs) and released preb- further to form MLBs, which were clearly separated from
odies (RPBs) (Alvarez and Steinbüchel, 2002) were the cytoplasm with an implied boundary layer. MLBs had
observed (Fig. 3D right panel and Fig. 3E). The PBs were no visible contact to the cytoplasm membrane (Fig. 3D).
visible as flat to spherical, relatively electron-dense struc- The mean thickness of cell wall of M. smegmatis-Rv3804c
tures close to the cytoplasmic membrane as described was thicker (13 nm) compared with that of the control
previously (Daniel et al., 2004). PBs did not show a clearly M. smegmatis mc2155 (8 nm) (Fig. 3C).

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1580 A. A. Elamin et al. 䊏

Fig. 2. Localization of Mycobacterium tuberculosis Ag85A in M. smegmatis mc2155. Localization was observed by electron microscopic
sections using a monoclonal antibody against Ag85A and post-embedding immunogold labelling (black dots). Ag85A was observed at inner
side of cell membrane, cell wall, in the cytoplasm and on the surface of TAG inclusions. PB, lipid prebody; RPB, released prebody; MLB,
matured lipid body; CW, cell wall; CM, cell membrane.

Lipid body development an approximate twofold increase of TDM and TMM in


M. smegmatis-Rv3804c compared with M. smegmatis
Ag85A overexpression led to an increased amount of lipid
mc2155 (Fig. 6A). This result is in agreement with the TLC
droplets in the cell. Using Bodipy 493/503 staining, the
results which show an increase of the glycolipids TMM and
application of flow cytometry allows the monitoring of lipid
TDM for M. smegmatis-Rv3804c (Fig. 6C). Expression of
droplet development. Bodipy 493/503 selectively stains
Ag85A modulates also to the composition and concen-
neutral lipids and is more specific for cellular lipid droplets
tration of mycolic acids. Upon HPLC analysis of their
than staining with Nile red (Gocze and Freeman, 1994).
mycolates, both strains have two slightly distinct
M. smegmatis mc2155 and M. smegmatis-Rv3804c cells
HPLC-double-cluster patterns (Fig. 6B). In comparison
were collected from the main culture at 4, 6, 8, 12, 16 h
with M. smegmatis mc2155 the double-cluster pattern from
and after induction (Figs 4 and 5). M. smegmatis-Rv3804c
M. smegmatis-Rv3804c shows a longer retention time on a
showed a higher fluorescence intensity (101–104) com-
C18 reverse-phase column, indicating the presence of
pared with M. smegmatis mc2155 (100–102), indicating
mycolic acids with longer carbon chains (Fig. 6B) (Wong
the presence of neutral lipid droplets in M. smegmatis-
et al., 1979; Butler et al., 1986; Cage, 1992). TLC results
Rv3804c (Raschke and Knorr, 2009). Fluorescence
indicate as well a slight increase of the mycolic acid con-
intensity of M. smegmatis-Rv3804c increased after 12 h
centration in the M. smegmatis-Rv3804c. The relative
(Fig. 4D–F) showing the increasing amount of neutral
amounts of a′-mycolic acids, a-mycolic acids and epoxy
lipids in the cells. M. smegmatis-mRv3804c harbouring the
mycolic acids increased (Fig. 6C).
plasmid pMV261::85A* (mutated Ag85A) was collected
Thus, Ag85A induces a quantitative and qualitative
at same time points and stained with Bodipy 493/503.
modulation of glycolipids and mycolic acids of the cell
M. smegmatis-mRv3804c strain showed very few lipid
wall. In addition we also observed an increase of storage
bodies in the cytoplasm in all time points (Fig. S2). Obvi-
lipids in the cell. M. smegmatis-Rv3804c shows a higher
ously M. smegmatis-mRv3804c showed changes in the
amount of TAGs in the TLC analysis (Fig. 6D).
cell size and morphology as M. smegmatis-Rv3804c but
without any effect on the lipid bodies formation.
Ag85A has DGAT activity
Ag85A modulates the synthesis of glycolipids, mycolic
The increase of TAGs in M. smegmatis-Rv3804c
acids and TAGs in M. smegmatis-Rv3804c
prompted us to ask the question whether Ag85A has an
In order to confirm that the altered morphology of acyltransferase activity required for TAG synthesis. TAGs
M. smegmatis-Rv3804c phenotype was also the cause of are synthesized by DGAT. DGAT enzymes mediate TAG
changes in cell envelope lipid composition, we analysed formations from long-chain acyl-coenzyme A (CoA) as
the lipid content of wild-type M. smegmatis and acyl donor and long-chain fatty alcohols or diacylglycerols
M. smegmatis-Rv3804c cells. We performed the total lipid as acyl acceptors (Wältermann et al., 2007).
analysis using our newly reported mycolyltransferase In order to test for DGAT activity, purified recombinant
assay and the equations (Elamin et al., 2009). The expres- M. tuberculosis Ag85A from Escherichia coli was assayed
sion of Ag85A led to an increase of concentration of with palmitoleoyl-CoA as acyl donor and 1,2-dipalmitoyl-
the glycolipids TDM and TMM in the cell. We detected sn-glycerol (1,2-dipalmitin) as the acyl acceptor. The

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1581

Fig. 3. Ag85A overexpression leads to lipid bodies formation and


changes the cell wall thickness.
A–D. Giant cells overexpressing Ag85A (right panel) from the heat
shock promoter phsp60 exhibit to numerous lipid bodies and a
thicker cell wall than wild type. The wild-type (M. smegmatis
mc2155) cells (left panel) displays normal shape, size with no lipid
bodies. Fig. 4. Monitoring of lipid body development. Flow cytometry
E. Schematic display of the stages of lipid body formation in using Bodipy 493/503 staining showed an increase of lipid bodies
M. smegmatis-Rv3804c (D, right panel). PB, lipid prebody; RPB, during the incubation time and induction for M. smegmatis-
released prebody; MLB, matured lipid body; CW, cell wall; CM, cell Rv3804c, while M. smegmatis mc2155 showed no change in the
membrane. fluorescence histogram. Cells were collected for FACS analysis at
(A) 4, (B) 6, (C) 8, (D) 12, (E) 16 h and (F) after induction.
© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592
1582 A. A. Elamin et al. 䊏

Fig. 5. Bodipy 493/503 staining giant cells overexpressing Ag85A from the heat shock promoter. Fluorescence images of
M. smegmatis-Rv3804c cells stained with Bodipy 493/503 and were collected at (A) 4, (B) 6, (C) 8, (D) 12, (E) 16 h and (F), (H) and (I) after
induction, M. smegmatis mc2155 used as control (G). The white arrows (A) indicated the starting of lipid accumulation and the arrows in (B)
indicated the lipid prebodies (PBs).

formation of the product TAG was verified by TLC (Fig. 7D Menten kinetics (Fig. 7A). For palmitoleoyl-CoA an
and E). The TLC results show clearly the production of a apparent Km of 390 mM was determined. Vmax was
band at the same Rf as the tripalmitin standard. This 3166 nmol mg-1 min-1 and Kcat of 55.54 min-1. The
product band was analysed by MALDI-TOF-MS and was Ser126Ala mutant did not show any detectable activity
verified as tripalmitin. When using the Ser126Ala mutant of (Fig. 7A).
Ag85A, no band corresponding to tripalmitin could be The Km values reported here for Ag85A are quite similar
detected, confirming Ser126 as the catalytic residue for to those reported for the M. tuberculosis DGAT TGS1
DGAT activity (data not shown). To determine the kinetic (Sirakova et al., 2006), an enzyme having preference for
parameters for Ag85A, the enzyme was assayed using an C26:0-CoA over C18:1-CoA with Km values 306 and
enzyme assay which quantifies the thiol groups of released 540 mM respectively. Another reported kinetic studies for
CoA-SH (Arabolaza et al., 2008) (see Experimental DGAT of M. tuberculosis TGS1–4 showed similar range of
procedures). Palmitoleoyl-CoA was used as acyl donor values by using diolein and oleoyl-CoA as substrates
in concentrations ranging from 0–2.5 mM and with 1,2- (Wältermann et al., 2007).
dipalmitoyl-sn-glycerol (1,2-dipalmitin) as the acyl accep- The molecular binding model (see below) predicted that
tor at a fixed concentration of 2 mM. The reaction curve 1,2-dipalmitin and palmitoleoyl-CoA can compete for
indicated that the DGAT reaction followed Michaelis– binding to the active site and elevated concentrations of

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1583

Fig. 6. Modulation of glycolipids, mycolic acids and TAGs in M. smegmatis-Rv3804c.


A. Quantification of TMM and TDM by the mycoltransferase assay.
B. HPLC trace of mycolic acid p-bromophenacyl esters from Mycobacterium smegmatis mc2155 and M. smegmatis-Rv3804c, showing a
double-cluster pattern. The pattern of M. smegmatis-Rv3804c has a longer retention time.
C and D. Total lipid analysis of different strains by analytical TLC. (C) Petroleum ether:diethyl ether:acetone (80:20:2, by vol.) was used as
mobile phase. M. smegmatis-Rv3804c shows stronger signal for glycolipids (TMM, TDM) and mycolic acids. Lane 1, M. smegmatis mc2155.
Lane 2, M. smegmatis-Rv3804c. (D) n-hexane:diethyl ether (9:1, by vol.) was used as mobile phase. M. smegmatis-Rv3804c has a stronger
signal for TAGs. Staining was performed with Coomassie blue as described (Elamin et al., 2009). Lane 1, M. smegmatis mc2155. Lane 2,
M. smegmatis-Rv3804c.

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1584 A. A. Elamin et al. 䊏

Fig. 7. DGAT activity of recombinant M. tuberculosis Ag85A and the Ser126Ala mutant.
A. Michaelis–Menten kinetics of Ag85A for palmitoleoyl-coenzyme A as substrate. The different substrate concentrations tested follow the
Michaelis–Menten equation (䊉). The Km and Vmax of palmitoleoyl-CoA were determined as 390 mM and 3166 nmol mg-1 min-1 respectively. The
mutant Ag85A (Ser126Ala) did not have any detectable activity ( ).
B and C. The apparent Km and Kcat of 1,2-dipalmitin for M. tuberculosis Ag85A were calculated at two concentrations of palmitoleoyl-coenzyme
A, 100 mM (B) and 700 mM (C). The Km and Kcat values for 1,2-dipalmitin were 9.09 mM and 38.57 min-1 respectively at palmitoleoyl-coenzyme
A concentration of 100 mM. At 700 mM of palmitoleoyl-coenzyme A the Km and Kcat values for 1,2-dipalmitin were 4.34 mM and 124.3 min-1
respectively.
D. TLC analysis of the extracted lipids from the reactions mixture. 1: Dipalmitin as reference, 2: Tripalmitin as reference, 3: Ag85A with 700 mM
palmitoleoyl-coenzyme without dipalmitin A, 4: Ag85A with 700 mM palmitoleoyl-coenzyme A and 10 mM dipalmitin, 5: Ag85A with 700 mM
palmitoleoyl-coenzyme A and 20 mM dipalmitin, 6: Ag85A with 700 mM palmitoleoyl-coenzyme A and 30 mM dipalmitin and 7: Ag85A with
700 mM palmitoleoyl-coenzyme A and 40 mM dipalmitin.
E. MALDI-TOF-MS analysis of the TLC spot obtained from Ag85A reaction, which was scratched from the TLC plates and extracted by
chloroform. The mass spectrum showed the formation of tripalmitin (arrow).
F. Model for inhibition of activity by dipalmitin at low palmitoleoyl-coenzyme A concentration. At high concentration of the acyl donor no
inhibitory effect is observed. Only at low palmitoleoyl-coenzyme A concentration dipalmitin may bind to the active site and block the entry of
the acyl donor.

1,2-dipalmitin should result in substrate inhibition. To test Substrate recognition


the model, we assayed enzyme activity at concentration
lower (100 mM) and at nearly double concentration of the Since the palmitoleoyl-CoA and 1,2-dipalmitin were
Km of palmitoleoyl-CoA (700 mM). The inhibitory effect of shown to be acyl donor and acyl acceptor, respectively,
1,2-dipalmitin at fixed palmitoleoyl-CoA concentration of we investigated the possible binding site of both sub-
100 mM is clear (Ki = 84 mM) (Fig. 7B). At 700 mM almost strates. The crystal structure of the homologous protein
no inhibition of 1,2-dipalmitin was observed (Ki = 6.8 mM). Ag85C has been determined (Ronning et al., 2000).
The Km for 1,2-dipalmitin was determined as 9.09 mM and Similar to Ag85C, Ag85A contains a catalytic triad formed
4.34 mM at palmitoleoyl-CoA concentrations of 100 and by residues Ser126, His262 and Glu230 and also pos-
700 mM respectively (Fig. 7B and C). sesses the highly conserved serine 126 in a deep
In summary, the experiments show clearly that Ag85A of trehalose binding groove as reported for Ag85B and C
M. tuberculosis exhibits DGAT activity and is an acyltrans- (Ronning et al., 2000; Anderson et al., 2001). The acyl
ferase involved in TAG biosynthesis. donor palmitoleoyl-CoA was docked to the trehalose

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1585

Fig. 8. Proposed catalytic mechanism of acyl transfer and hypothetical binding mode of acyl substrates.
Upper panel. Schematic representation of the proposed catalytic mechanism of acyl transfer. The reaction comprises three-steps: (1) Ser126
allows a nucleophilic attack on the carboxylate carbon of the acyl donor palmitoleoyl-coenzyme A, leading to an acyl-enzyme adduct. (2) The
hydroxyl group of the acyl acceptor 1,2-dipalmitoyl-sn-glycerol performs a nucleophilic attack on the carboxylate carbon of the acyl-enzyme
adduct. (3) The triacylglycerol product is released. The transferred palmitoleoyl moiety is in red.
Lower panel. Proposed binding mode for the acyl substrates. (A) Docked palmitoleoyl-coenzyme A in the binding pocket of antigen 85A. The
thioester carbonyl group is oriented in the vicinity of Ser126 (arrow). Carbon atoms of the docked palmitoleoyl-coenzyme A molecule are
green. (B) Proposed intermediate with the palmitoleic acid covalently attached to Ser126. (C) Complex of the Ag85-palmitoleoyl adduct with
1,2-dipalmitoyl-sn-glycerol. Carbon atoms of the 1,2-dipalmitoyl-sn-glycerol are in grey. The figure shows that the palmitoyl chains are
positioned on top of the palmitoleoyl-Coenzyme A molecule and may block the entry of the acyl donor at high 1,2-dipalmitin concentration. The
position of the active-site Ser126 is indicated with an asterisk. The figure was prepared with Pymol (http://www.pymol.org/).

binding pocket with affinities of -9.3 kcal mol-1. The C) synthesize the most abundant glycolipid of the myco-
thioester carbonyl group is located in close proximity to bacterial cell wall, the cord factor. The cord factor (TDM)
the serine 126 and allows a nucleophilic attack by the is essential for the integrity of the mycobacterial cell wall
serine hydroxyl group leading to the proposed acyl- and pathogenesis (Nguyen et al., 2005). The enzymes
enzyme adduct (Ronning et al., 2000). The long palmito- of the antigen 85 complex may possess as well the
leoyl chain occupies the deep hydrophobic groove on capability to confuse and evade the host immune
the protein as observed for palmitoyl in thioesterase 1 system (Armitige et al., 2000; Harth et al., 2002;
(Fig. 8A) (Bellizzi et al., 2000). The acyl acceptor 1,2- Ronning et al., 2004).
dipalmitin was docked with an affinity of 4.7 kcal mol-1 In order to study the quantitative effect of Ag85A on the
to the assumed acyl-enzyme adduct (Fig. 8B). 1,2- of mycobacterial cell wall synthesis, we expressed the
Dipalmitin accommodates the same space in the binding M. tuberculosis Ag85A in the non-pathogenic M. smegma-
pocket as trehalose in Ag85B (Anderson et al., 2001). The tis mc2155 which was used as a model organism because
palmitoyl chains cover the binding site of palmitoleoyl- it is fast growing and shares many features with pathogenic
CoA and may block the entry of the acyl donor at high mycobacteria (He and De Buck, 2010). The recombin-
1,2-dipalmitin concentration. The pantoic acid component ant M. smegmatis-Rv3804c expressing M. tuberculosis
of CoA and an ester group of 1,2-dipalmitin adapts the Ag85A displayed thickening of the cell wall and major
glucose ring structure of trehalose bound to Ag85B (PDB changes in morphology. Unexpectedly, in addition to the
1F0P) and the head region of the palmitoleoyl chain occu- morphological changes, an extensive formation of lipid
pies approximately the same position as MPD bound to bodies was observed. The presence of neutral and
Ag85B (Anderson et al., 2001) (data not shown). saturated TAGs which are a major components of lipid
inclusions (Daniel et al., 2004; Sirakova et al., 2006; Wäl-
termann et al., 2007) were confirmed by Bodipy 493/503
Discussion staining. TAGs are the major storage molecules for eukary-
The enzymes of the antigen 85 complex (Ag85A, B and otic organisms (Yen et al., 2005; 2008; Saha et al., 2006)

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1586 A. A. Elamin et al. 䊏

but these are not common storage compounds of bacteria (isolated from lipid bodies) (Lardizabal et al., 2001) and
(Alvarez et al., 2000). Only bacteria belonging to the His-Sco0958 DGAT from Streptomyces coelicolor. (Arabo-
genera Acinetobacter (Kalscheuer and Steinbüchel, laza et al., 2008). The enzyme exhibits a high Km of 390 mM
2003), Streptomyces (Olukoshi and Packter, 1994; Arabo- as reported previously for M. tuberculosis TGS1 (Sirakova
laza et al., 2008), Rhodococcus (Kalscheuer and Stein- et al., 2006).
büchel, 2003) and Mycobacterium (Daniel et al., 2004; The known Acyl-CoA:DGAT such as DGAT1 or
Sirakova et al., 2006; Low et al., 2010) accumulate TAGs. DGAT2 belong to two distinct protein families, have dif-
We found that the recombinant Ag85A was partly ferent physiological roles and exhibit no sequence
membrane-bound and also localized in the cytoplasm or on homology to each other (Kalscheuer and Steinbüchel,
surface of lipid inclusions in the recombinant M. smegma- 2003; Yen et al., 2005; 2008). Ag85A does not belong to
tis strain as observed for other lipid-synthesizing enzymes either of the two known DGAT families. It does not
such as the 14 kDa phasin from Rhodococcus ruber contain putative DGAT motifs, which have been pro-
and phasin PhaP1 from Ralstonia eutropha (Pieper-Fürst posed by Liu et al. (2010) or Kalscheuer and Stein-
et al., 1995). büchel (2003). The DGAT activity of Ag85A includes two
Flow cytometry revealed a major difference in stain- consecutive reactions: fatty acyl-CoA hydrolysis (thio-
ing pattern between M. smegmatis mc2155 cells and esterification), and transfer of the acyl group to the dia-
M. smegmatis-Rv3804c which contained very large lipid cylglycerol (transesterification). Ag85A shares these two
bodies. An increase in lipid droplet size led to an catalytic activities with other serine hydrolases such as
increase in fluorescence intensity allowing a qualitative thioesterases (Bellizzi et al., 2000; Cao et al., 2009;
statement about the lipid droplet development. The con- Dias et al., 2010) and carboxylesterases (Satoh and
stantly observed low fluorescence in wild type could Hosokawa, 1998; Bencharit et al., 2006). Thioesterases
result from the fact that the mycobacterial cells contain exhibit only acyl-CoA hydrolase activity, while some
different types of neutral lipids. Comparing FACS infor- eukaryotic carboxylesterases also exhibit additional acyl-
mation with the cell images provided by fluorescence transferase (ACAT) activity like human carboxylesterase
microscope, we conclude that the increase of fluores- 1 (Satoh and Hosokawa, 1998; Bencharit et al., 2006).
cence is caused by the increased size of neutral lipid Thioesterases and carboxylesterases share a catalytic
droplets. The TEM and fluorescence staining experi- mechanism where a conserved serine hydrolyses the
ments showed that lipid biosynthesis begins at periph- substrate by a nucleophilic attack. The Ag85A homo-
eral lipid domains close to the cytoplasm membrane logue Ag85C is proposed to act as a serine hydrolase
followed by the subsequent formation of PBs. The exhibiting mycolyl esterase/transferase activity. Ag85A
spherical PBs enlarge and are released from the contains the same catalytic triad as Ag85C formed by
membrane forming prebodies followed by full release residues Ser126, His262 and Glu230 and possesses a
into the cytoplasm to form mature lipid bodies deep substrate binding groove near the active-site
(Wältermann et al., 2005). It is difficult to distinguish serine as reported for Ag85B and C (Ronning et al.,
between MLBs from RPBs but the former were larger 2000; Anderson et al., 2001). The positions of the sub-
and clearly separated from the cytoplasm. The control strates in the docking model suggest a similar mecha-
(parental) M. smegmatis mc2155 strain (Fig. S1) and nism for Ag85A as proposed for Ag85C (Ronning et al.,
M. smegmatis-mRv3804c, the later contains the mutated 2000). The active-site Ser126, verified by the analysis of
Ag85A, showed no or very rarely few lipid bodies in the a Ser126Ala mutant, allows a nucleophilic attack by the
cytoplasm. Interestingly, M. smegmatis-mRv3804c dis- serine hydroxyl group on the carboxylate carbon of the
played the same large cell phenotype which was fatty acyl-coenzyme, forming an acyl-enzyme intermedi-
observed in M. smegmatis-Rv3804c, but without induc- ate. Finally the short distance of the hydroxyl group of
tion of the lipid bodies (Fig. S2) supporting the possibility the acyl acceptor diacylglycerol would permit a nucleo-
that the Ag85A may had DGAT activity. philic attack on the carboxylate carbon of the acyl-
Acyl-CoA:DGAT catalyse the biosynthesis of TAGs enzyme adduct. These findings, establish for the first
(Kalscheuer and Steinbüchel, 2003). The accumulation of time a link between cell wall and TAG biosynthesis,
neutral lipids by Ag85A transformants of M. smegmatis which will shed more light and expand our understand-
mc2155 prompted us to examine Ag85A for Acyl- ing of the role of Ag85A in M. tuberculosis pathogenesis
CoA:DGAT activity. We found that Ag85A mediates TAG and persistence. Since the current data do not comprise
formation from long-chain acyl-CoA as acyl donor and final proof of Ag85A involvement in TAG synthesis under
1,2-dipalmitoyl-sn-glycerol (1,2-dipalmitin) as the acyl dormant conditions in M. tuberculosis we have initiated
acceptor (Fig. 7) as is known for DGAT enzymes from other further work on construction of suitable k.o. mutants and
organisms (Wältermann et al., 2007). The DGAT activity of their detailed characterization under various growth con-
Ag85A is in the same range as is reported for fungal DGAT2 ditions including dormancy.

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1587

Experimental procedures introduction of single specific mutations into Ag85A to


mutated serine 126 to alanine (tcg to gcg) was achieved by
Gene cloning and transformation into M. smegmatis the use of pET28::Ag85A and pMV261::85A as templates
mc2155 DNA together with the following pair of mutagenic primers:
forward 5′-gtc gtc ggt ctt gcg atg gct gct tc-3′ and reverse
The gene coding for Ag85A (FbpA; Rv3804c) was amplified by 5′-gaa gca gcc atc gca aga ccg acg ac-3′ (the nucleotide
PCR using genomic DNA from M. tuberculosis H37Rv as changes giving the appropriate mutations are shown in
template. The sequence of PCR sense primer for Rv3804c bold). After initial denaturation at 95°C for 0.5 min, the
was 5′-ggt acc gga tcc atg cag ctt gtt gac agg gt-3′ and that of cycling parameters were 0.5 min at 95°C followed by 1 min
anti-sense primer was 5′-ggc tgt atc gat cta gtg atg gtg atg gtg at 55°C and 7 min at 68°C (16 cycles). The reaction mix-
atg ggc gcc ctg ggg cgc ggg cc-3′. The PCR products were tures were placed on ice for 2 min, and then the parental,
purified by agarose gel-electrophoresis followed by extraction supercoiled double-stranded DNA was digested with 1 ml of
using the QIAquick kit (Qiagen, Germany). After cleavage with DpnI at 37°C for 1 h before being transformed into compe-
restriction endonucleases FastDigest BamHI and FastDigest tent E. coli Top10 F⬘. Mutation was verified by DNA
ClaI (Fermentas) fragments were purified and ligated in sequencing. The expression and purification of pET28::85A*
pMV261 (Stover et al., 1991), digested with the same restric- (mutated 85A) was done by using BL21 (DE3) cells
tion endonucleases to generate plasmid pMV261::85A. The (Novagen) as described (Elamin et al., 2009). The isolation
ligation mixture was used to transform electrocompetent cells and transformation of pMV261::85A* (mutated Ag85A) into
of E. coli Top10 F⬘ (Invitrogen). The cells were grown in M. smegmatis mc2155 was done as described above. The
Luria–Bertani (LB) broth and LB agar supplemented with M. smegmatis strain mc2155, harbouring the plasmid
kanamycin (20 mg ml-1) and incubated at 37°C. pMV261::85A pMV261::85A* was named M. smegmatis-mRv3804c.
was isolated from transformed colonies and plasmid integrity
was confirmed by DNA sequencing (Department of Genom-
analytik, HZI Braunschweig). For transformation into Expression of Ag85A in M. smegmatis-Rv3804c and
M. smegmatis mc2155 pMV261::85A was isolated using the M. smegmatis-mRv3804c
QIAprep Spin Miniprep Kit (Qiagen, Germany). Preparation
and transformation of M. smegmatis mc2155 was carried out Mycobacterium smegmatis mc2155 was grown in 7H9
as described before (Pelicic et al., 1996) with minor modifica- medium (Difco) supplemented with ADC, 0.2% glycerol and
tions. The resulting suspensions from electroporation were 0.05% Tween 80. Induction of the recombinant gene was
incubated at 37°C for 4 h and then plated on kanamycin done by shifting the incubation temperature from 37 to 45
(20 mg ml-1)-containing plates (Middlebrook 7H10 agar degrees for 2 h if not otherwise stated.
supplemented with ADC, 0.2% glycerol and 0.05% Tween 80.
Transformed colonies were confirmed by PCR amplification
and DNA sequencing. The M. smegmatis strain mc2155, har- Preparation for field emission scanning electron
bouring the plasmid pMV261::85A was named M. smegmatis- microscopy (FESEM)
Rv3804c.
Mycobacterium smegmatis mc2155, M. smegmatis-Rv3804c
Gene cloning, transformation into E. coli BL21 and and M. smegmatis mc2155-pMV261 were grown at 37°C for
production of Ag85A 48 h in 75 ml flasks containing 25 ml of Middlebrook 7H9
broth, supplemented with ADC, 0.2% glycerol and 0.05%
For kinetic analysis, Ag85A was produced in large amounts Tween 80 and kanamycin (20 mg ml-1) (except for M. smeg-
as recombinant 6¥His-tagged protein. The gene encoding the matis mc2155) and diluted to obtain 103–104 cells ml-1. The
secreted form of antigen 85A (FbpA; Rv3804c) was amplified diluted suspensions were added to 100 ml flasks containing
by PCR. The sequence of PCR sense primer for Rv3804c Middlebrook 7H9 broth until an OD600 of 0.8–1 was reached,
was 5′-ggc tgt cat atg gca ttt tcc cgg ccg ggc tt-3′ and that of and then the cultures were incubated at 45°C for additional
anti-sense primer was 5′-ggt acc gga tcc cta ggc gcc ctg ggg 2 h. Samples of 5 ml were fixed with a solution containing 2%
cgc gg-3′. The PCR product was digested with FastDigest glutaraldehyde and 5% formaldehyde in cacodylate buffer
NdeI and FastDigest BamHI (Fermentas). The agarose-gel- (0.1 M cacodylate, 0.09 M sucrose, 0.01 M MgCl2, 0.01 M
purified DNA was ligated with T4 DNA ligase (Roche) in CaCl2, pH 6.9) for 1 h on ice and washed with TE buffer
double-digested pET-28a (+) (Novagen). The final plasmid (20 mM TRIS, 2 mM EDTA, pH 6.9). After washing with TE
pET28::Ag85A contained the DNA sequence coding for buffer samples placed onto poly-l-lysine-coated slides and
antigen 85A with an N-terminal 6¥histidine tag. The expres- then dehydrated with 10%, 30%, 50%, 70%, 90%, 100%
sion plasmid pAg85A was transformed into BL21 (DE3) cells acetone, each step for 10 min on ice. The last 100% acetone
(Novagen). The correct orientation and integrity of inserts step was performed at room temperature. Samples were then
were confirmed by PCR amplification and DNA sequence critical-point-dried with liquid CO2 (CPD 030, Bal-Tec, Liecht-
analysis. BL21 (DE3) cells containing pET28::Ag85A were enstein) mounted onto aluminium stubs and sputter coated
cultivated and the recombinant protein was purified as with gold (SCD 500, Bal-Tec, Liechtenstein) before examina-
reported before (Elamin et al., 2009). tion in a Zeiss field emission scanning electron microscope
Site-directed mutagenesis Gemini DSM852 (Zeiss, Oberkochen, Germany) at an accel-
eration voltage of 5 kV using the Everhart-Thornley SE detec-
Site-directed mutagenesis was performed by use of a tor and the inlens SE detector in a 50:50 ratio. Images are
QuikChange site-directed mutagenesis kit (Stratagene). The stored onto a 230 MB MO disk.

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1588 A. A. Elamin et al. 䊏

Transmission electron microscopy millilitre of M. smegmatis mc2155, M. smegmatis-Rv3804c


and M. smegmatis-mRv3804c cultures which were grown as
The same collected samples as described above were fixed described were centrifuged, washed with PBS and resus-
in a fixation solution containing 5% formaldehyde and 2% pended in 90 ml of PBS. Ten microlitres of a 1 mg ml-1 solution
glutaraldyhde in cacodylate buffer (0.1 M cacodylate, 0.01 M of Bodipy 493/503 (Invitrogen) in ethanol was added and the
CaCl2, 0.01 M MgCl2, 0.09 M sucrose, pH 6.9) and washed bacteria were incubated for 10 min in the dark at room
with cacodylate buffer. Samples were then osmificated with temperature. After washing twice with PBS 2 ml of the Bodipy-
1% aqueous osmium for 1 h at room temperature, washed labelled bacteria were put onto a glass slide, covered with a
and pellets of the samples were embedded in 2% water coverslip and examined using a Zeiss photomicroscope with
agar and cut into small cubes. Dehydration was achieved an attached Hrc camera and Axiovision software 4.7. For time
with a graded series of acetone (10%, 20% and 50%) for collection experiments the samples were collected from the
30 min on ice followed by contrasting with 2% uranyl acetate main culture at 4, 6, 8, 12 and 16 h and also after 2 h induc-
in 70% acetone for overnight at 4°C and further dehydrated tion at 45°C.
with 90% and 100% acetone. Samples in the 100% acetone
step were allowed to reach room temperature and were infil-
trated with the epoxy resin according to Spurr’s formula for Flow cytometry
a medium resin: 1 part 100% acetone/1 part resin for over-
night, 1 part 100% acetone/2 parts resin for 8 h, pure resin Depending on given cell density, the cell number used for
for overnight and several changes the following 2 days. measurement was adjusted leading to 700–1500 events per
Samples were then transferred to resin filled gelatine cap- second. Cell suspension volume used was 100 ml and the
sules and polymerized for 8 h at 75°C. Ultrathin sections flow rate was adjusted. The PI (Invitrogen) staining was
were cut with a diamond knife, picked up with formvar- used to determine the vitality. PI was added to gain a final
coated copper grids (300 mesh) and counterstained with 2% concentration of 15 mM. The sample was incubated for
aqueous uranyl acetate and lead citrate. After air-drying 10 min and directly measured. The lipid bodies staining
samples were examined in a Zeiss transmission electron were performed using Bodipy 493/503 (Invitrogen) as
microscope TEM910 at an acceleration voltage of 80 kV. described previously. Flow cytometry was conducted using
Images were recorded digitally with a Slow-Scan CCD- a BD FACS Canto II (BD Biosciences) equipped with an
Camera (ProScan, 1024 ¥ 1024, Scheuring, Germany) with air-cooled 15 mW 500 nm Argon-Laser. For both PI and
ITEM Software (Olympus Soft Imaging Solutions, Münster, Bodipy 493/503 staining the same settings were used. Flow
Germany). cytometry data were analysed using the BD FACSDiva Soft-
ware (BD Biosciences).

Immunoelectron microscopic labelling Mycolic acids extraction and derivatization procedures


Mycobacterium smegmatis mc2155 and M. smegmatis- Mycobacterial cultures (0.5 ml) (M. smegmatis mc2155 and
Rv3804c cells which treated as described were fixed with 1% M. smegmatis-Rv3804c) were harvested, washed and resus-
formaldehyde directly in the growth medium for 2 h at 4°C, pended in 2 ml of saponification reagent (25% KOH in 50%
washed twice with cacodylate buffer containing 10 mM ethanol) per gram whole cells followed by saponification by
glycine. Samples were dehydrated with a graded series of autoclaving for 1 h at 121°C. Saponified cells were cooled to
ethanol (10%, 30%, 50%, 70%, 90% and 100%) and embed- room temperature and acidified to pH 1 with 6 N HCI. The
ded in LRWhite resin. Ultrathin sections were cut with a mycolic acids were extracted by adding 2 ml of chloroform
diamond knife and collected onto formvar coated nickel grids per gram cells, mixed in a separation funnel to assist sepa-
(300 mesh). Sections were incubated with mouse anti-85A ration of the chloroform layer. The extracted mycolic acids
mAb IgG (Lionex, Germany) for 12 h at 4°C (1:5 dilution). were evaporated to dryness and resuspended in chloroform
After washing with PBS bound antibodies were made visible for derivatization. The mycolic acids were derivatized to
with protein A gold complexes (15 nm in diameter, 1:200 p-bromophenacyl esters by the method of Durst et al. (1975)
dilution of the stock solution, incubation for 1 h at room by p-bromophenacyl-8 reagent (Pierce Chemical). Derivati-
temperature). After washing in PBS and distilled water sec- zation was carried out for 45 min at 85°C in glass tubes. After
tions were counterstained with 4% aqueous uranyl acetate derivatization, the p-bromophenacyl esters of the mycolic
for 2 min before examination in a Zeiss transmission electron acids were filtered through 0.45-mm-pore nylon 66 membrane
microscope EM910 at an acceleration voltage of 80 kV and filters, evaporated to dryness and resuspended in chloroform
calibrated magnifications. Images were recorded digitally for analysis.
with a Slow-Scan CCD-Camera (ProScan, 1024 ¥ 1024,
Scheuring, Germany) with ITEM-Software (Olympus Soft
Imaging Solutions, Münster, Germany). HPLC conditions

HPLC of the derivatized mycolic acids was performed with an


Bodipy labelling HPLC model 450 data system controller (KNAUER’s Smart-
line, Germany). The UV light absorbing p-bromophenacyl
Bodipy dyes have potential applications as stains for neutral esters were detected with a variable-wavelength detector
lipids in eukarotic as well as in prokaryotic cells for staining (Smartline UV Detector 2500) set at 254 nm. A reverse-phase
intracellular lipid droplets (Gocze and Freeman, 1994). One C-18 cartridge column (22 cm by 2.1 mm; Pierce) was used

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1589

to separate the mycolic acid esters. A solvent gradient system acid. 100 ml of the supernatant was taken after centrifugation
of chloroform and methanol was controlled by two Smartline (17 000 g for 10 min), and added to 100 ml of 2 mM DTNB
Pump 1000 solvent delivery pumps (KNAUER) adjusted dissolved in Tris-HCl (pH 7.8) in a F-bottom 96-well plates
to a constant flow rate of 0.6 ml min-1. The column was (Greiner Bio-One). The product formation was measured by
equilibrated to 10% chloroform-90% methanol over a 13 min reading the absorbance at 405 nm with a microplate reader
period. After the injection of 5 ml of samples, the solvent MRX Revelation (Dynex Technologies GmbH, Germany).
concentration was changed linearly over a 1 min period to The rate of product formation (nmol mg-1 min-1) was calcu-
25% chloroform-75% methanol; over the next 30 min period, lated using a standard curve of acetylcysteine from 0 to
the solvent concentration was changed linearly to 70% 100 mM. Kinetic data were calculated with GraphPad Prism
chloroform-30% methanol. software using the Michaelis–Menten model (GraphPad Soft-
The HPLC fractions and total lipids were analysed by ware, La Jolla, CA, USA).
thin-layer chromatography (TLC) on silica plates (Macherey- The apparent Km 1,2-dipalmitin for M. tuberculosis Ag85A
Nagel, Germany) with the different solvents system. For ana- was determined at two concentrations of palmitoleoyl-CoA,
lytical purposes, the spots were visualized with Coomassie 100 mM and 700 mM. The product rate was calculated as
blue. Each developed TLC plate was air dried and immersed mentioned above. In order to verify the acylation of diacylg-
in a staining solution consisting of 0.04% Brilliant blue R lycerol by Ag85A to TAG, the assay was performed with
(Sigma) in 25% methanol. After 30 min, the plate was the same conditions as mentioned before for Ag85A (the
removed from the staining solution, immersed in 25% metha- protein final concentration increased to 12 mM) with 700 mM
nol (destaining solution), and allowed to stand for 2–5 min palmitoleoyl-CoA and different concentration of 1,2-
with occasional agitation. Then the plate was removed from dipalmitin (0–2000 mM). The reaction mixture was incubated
the destaining solution and excess liquid on the surface of the at 37°C for 2 h, and stopped and extracted by the addition
plate was soaked up with filter paper by gentle pressing. The of 1 ml of chloroform with gentle agitation for 6–12 h. The
box density was measured by Quantity one program (version mixture was allowed to stand for 15 min followed by sepa-
4.6.2) (Bio-Rad Laboratories). ration of the chloroform layer. The extracted lipids were
dried and resuspended in chloroform. The lipids were analy-
sed by TLC with n-hexane: Acetic acid (9:1, by vol.) as
Total lipids extraction and quantification of TMM mobile phase. Dipalmitin and tripalmitin (Sigma-Aldrich)
and TDM were used as references. Staining was performed with Coo-
massie blue in methanol as described before. The TLC spot
Bacterial cultures (0.5 l) from M. smegmatis mc2155 and corresponding to tripalmitin was scratched from the TLC
M. smegmatis-Rv3804c were used for total lipids extraction plates and extracted by chloroform and analysed by MALDI-
as described previously (Elamin et al., 2009). Quantification TOF-MS.
of TMM and TDM from the total lipid of mycobacterium cells
was performed by the mycolyltransferase assay as described
earlier (Elamin et al., 2009). The final TMM and TDM concen- Docking
tration was obtained by subtraction of the glucose concen-
tration of the negative control without antigen 85A from the Co-ordinates for the composition of the substrates were
glucose concentration obtained by the reaction. obtained by the HIC up server (http://xray.bmc.uu.se/hicup/).
Palmitoleic acid was extracted from the PDB entry 1FK3.
Dipalmitoyl-sn-glycero-3-phosphate was extracted from the
Determination of DGAT activity with purified PDB entry 1HG4. Acetyl coenzyme was extracted from 3F5O
recombinant antigen 85A (Cao et al., 2009). The substrates palmitoleoyl-CoA and
1,2-dipalmitin were constructed using the program Coot
The DGAT activity was assayed for purified recombinant pro- (Emsley and Cowtan, 2004). Energy minimization, addition
teins from BL21 (DE3) (Ag85A and mutated Ag85A) by using of full charges and polar hydrogens to the substrate atoms
5,5-dithiobis (2-nitrobenzoic acid) (DTNB), which specifically were performed by the Dundee PRODRG2 Server (http://
reacts with thiol groups of released CoA-SH (Arabolaza et al., davapc1.bioch.dundee.ac.uk/prodrg/) (Schüttelkopf and
2008). For the determination of kinetic constants of Ag85A van Aalten, 2004). The co-ordinates of Ag85A were obtained
with palmitoleoyl-CoA as substrate the final concentration of from the PDB entry 1SFR. AutoDock Tools (ADT) (http://
palmitoleoyl-CoA was varied between 25 and 2000 mM while autodock.scripps.edu/resources/adt) (Sanner, 1999) was
the concentration of 1,2-dipalmitin was kept constant at used for the preparation of the pdbqt files (addition of hydro-
2 mM. Stock solution of palmitoleoyl-CoA was dissolved in gens to the macromolecule and definition of rotatable torsion
CHAPS (5 mM). 1,2-Dipalmitin was suspended in CHAPS angles for the ligand). The ligand palmitoleoyl-CoA was
(40 mM) and solubilized by adding drops of chloroform/ docked within a search area defined by a grid box of size
methanol (1:1 by vol.). x = 32, y = 24 and z = 16 with a spacing of 1.0 Å and centred
The reaction was carried out in a buffer containing 100 mM at x = 41.38, y = 62.42, z = 3.10, encompassing the trehalose
potassium phosphate buffer (pH 7.0), 5% (v/v) ethanol and binding pocket.
10 mM MgCl2. The assay was performed in a total volume of Vina (Trott and Olson, 2010) was used for molecular
260 ml at 37°C. The reaction was initiated by adding pure docking. The hypothetical palmitoleoyl-Ag85A adduct was
Ag85A (57 mg) to the mixture (6 mM in final concentration). constructed with Coot by moving the molecule slightly so that
After 1 min an aliquot of 60 ml was taken and the reaction was one carbonyl-oxygen was near to the oxygen of serine126 to
stopped by the addition of 60 ml of 1% (w/v) trichloroacetic allow binding. 1,2-Dipalmitin was docked to the palmitoleoyl-

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1590 A. A. Elamin et al. 䊏

Ag85A complex by Vina with the following parameters: grid Butler, W.R., Ahearn, D.G., and Kilburn, J.O. (1986) High–
area x = 16, y = 26 and z = 16 with a spacing of 1.0 Å, performance liquid chromatography of mycolic acids as a
centred at x = 45.40, y = 68.016, z = 3.48. The program was tool in the identification of Corynebacterium, Nocardia,
run with an exhaustiveness of 20. Rhodococcus, and Mycobacterium species. J Clin Micro-
biol 23: 182–185.
Acknowledgements Cage, G.D. (1992) High-performance liquid chromatography
patterns of Mycobacterium gordonae mycolic acids. J Clin
We thank our colleagues, Wulf Oehlmann, Manfred Nimtz and Microbiol 30: 2402–2407.
Ina Schleicher, for their help and technical support. Ayssar Cao, J., Xu, H., Zhao, H., Gong, W., and Dunaway-Mariano,
Elamin is indebted to the Deutscher Akademischer Austaus- D. (2009) The mechanisms of human hotdog-fold
chdienst (DAAD) for the award of doctoral scholarship. thioesterase 2 (hTHEM2) substrate recognition and cataly-
sis illuminated by a structure and function based analysis.
References Biochemistry 48: 1293–1304.
Cardona, P.J. (2007) New insights on the nature of latent
Alvarez, H.M., and Steinbüchel, A. (2002) Triacylglycerols in tuberculosis infection and its treatment. Inflamm Allergy
prokaryotic microorganisms. Appl Microbiol Biotechnol 60: Drug Targets 6: 27–39.
367–376. Casadevall, A., and Pirofski, L.A. (2000) Host-pathogen inter-
Alvarez, H.M., Kalscheuer, R., and Steinbüchel, A. (2000) actions: basic concepts of microbial commensalism, coloni-
Accumulation and mobilization of storage lipids by Rhodo- zation, infection, and disease. Infect Immun 68: 6511–6518.
coccus opacus PD630 and Rhodococcus ruber NCIMB Coker, R.J. (2004) Review: multidrug-resistant tuberculosis:
40126. Appl Microbiol Biotechnol 54: 218–223. public health challenges. Trop Med Int Health 9: 25–40.
Anderson, D.H., Harth, G., Horwitz, M.A., and Eisenberg, D. Daffe, M., and Draper, P. (1998) The envelope layers of
(2001) An interfacial mechanism and a class of inhibitors mycobacteria with reference to their pathogenicity. Adv
inferred from two crystal structures of the Mycobacterium Microb Physiol 39: 131–203.
tuberculosis 30 kDa major secretory protein (Antigen 85B), Daniel, J., Deb, C., Dubey, V.S., Sirakova, T.D., Abomoelak,
a mycolyl transferase. J Mol Biol 307: 671–681. B., Morbidoni, H.R., and Kolattukudy, P.E. (2004) Induction
Arabolaza, A., Rodriguez, E., Altabe, S., Alvarez, H., and of a novel class of diacylglycerol acyltransferases and tria-
Gramajo, H. (2008) Multiple pathways for triacylglycerol cylglycerol accumulation in Mycobacterium tuberculosis as
biosynthesis in Streptomyces coelicolor. Appl Environ it goes into a dormancy-like state in culture. J Bacteriol
Microbiol 74: 2573–2582. 186: 5017–5030.
Armitige, L.Y., Jagannath, C., Wanger, A.R., and Norris, S.J. Dias, M.V., Huang, F., Chirgadze, D.Y., Tosin, M., Spiteller,
(2000) Disruption of the genes encoding antigen 85A and D., Dry, E.F., et al. (2010) Structural basis for the activity
antigen 85B of Mycobacterium tuberculosis H37Rv: effect and substrate specificity of fluoroacetyl-CoA thioesterase
on growth in culture and in macrophages. Infect Immun 68: FlK. J Biol Chem 285: 22495–22504.
767–778. Ducati, R.G., Ruffino-Netto, A., Basso, L.A., and Santos, D.S.
Bass, J.B., Jr, Farer, L.S., Hopewell, P.C., O’Brien, R., (2006) The resumption of consumption – a review on
Jacobs, R.F., Ruben, F., et al. (1994) Treatment of tuber- tuberculosis. Mem Inst Oswaldo Cruz 101: 697–714.
culosis and tuberculosis infection in adults and children. Durst, H.D., Milano, M., Kikta, E.J., Jr, Connelly, S.A., and
American Thoracic Society and The Centers for Disease Grushka, E. (1975) Phenacyl esters of fatty acids via crown
Control and Prevention. Am J Respir Crit Care Med 149: ether catalysts for enhanced ultraviolet detection in liquid
1359–1374. chromatography. Anal Chem 47: 1797–1801.
Beatty, W.L., and Russell, D.G. (2000) Identification of myco- Elamin, A.A., Stehr, M., Oehlmann, W., and Singh, M. (2009)
bacterial surface proteins released into subcellular com- The mycolyltransferase 85A, a putative drug target of
partments of infected macrophages. Infect Immun 68: Mycobacterium tuberculosis: development of a novel
6997–7002. assay and quantification of glycolipid-status of the myco-
Belisle, J.T., Vissa, V.D., Sievert, T., Takayama, K., Brennan, bacterial cell wall. J Microbiol Methods 79: 358–363.
P.J., and Besra, G.S. (1997) Role of the major antigen Emsley, P., and Cowtan, K. (2004) Coot: model-building tools
of Mycobacterium tuberculosis in cell wall biogenesis. for molecular graphics. Acta Crystallogr D Biol Crystallogr
Science 276: 1420–1422. 60: 2126–2132.
Bellizzi, J.J., 3rd, Widom, J., Kemp, C., Lu, J.Y., Das, A.K., Garton, N.J., Christensen, H., Minnikin, D.E., Adegbola, R.A.,
Hofmann, S.L., and Clardy, J. (2000) The crystal structure and Barer, M.R. (2002) Intracellular lipophilic inclusions of
of palmitoyl protein thioesterase 1 and the molecular basis mycobacteria in vitro and in sputum. Microbiology 148:
of infantile neuronal ceroid lipofuscinosis. Proc Natl Acad 2951–2958.
Sci USA 97: 4573–4578. Garton, N.J., Waddell, S.J., Sherratt, A.L., Lee, S.M., Smith,
Bencharit, S., Edwards, C.C., Morton, C.L., Howard-Williams, R.J., Senner, C., et al. (2008) Cytological and transcript
E.L., Kuhn, P., Potter, P.M., and Redinbo, M.R. (2006) analyses reveal fat and lazy persister-like bacilli in tuber-
Multisite promiscuity in the processing of endogenous culous sputum. PLoS Med 5: e75.
substrates by human carboxylesterase 1. J Mol Biol 363: Gocze, P.M., and Freeman, D.A. (1994) Factors underlying
201–214. the variability of lipid droplet fluorescence in MA-10 Leydig
Brennan, P.J., and Nikaido, H. (1995) The envelope of tumor cells. Cytometry 17: 151–158.
mycobacteria. Annu Rev Biochem 64: 29–63. Harth, G., Horwitz, M.A., Tabatadze, D., and Zamecnik, P.C.

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


Diacylglycerol acyltransferase activity of Ag85A 1591

(2002) Targeting the Mycobacterium tuberculosis 30/32- Pelicic, V., Reyrat, J.M., and Gicquel, B. (1996) Generation
kDa mycolyl transferase complex as a therapeutic strategy of unmarked directed mutations in mycobacteria, using
against tuberculosis: proof of principle by using antisense sucrose counter-selectable suicide vectors. Mol Microbiol
technology. Proc Natl Acad Sci USA 99: 15614–15619. 20: 919–925.
Epub 12002 Nov 15611. Pieper-Fürst, U., Madkour, M.H., Mayer, F., and Steinbüchel,
He, Z., and De Buck, J. (2010) Cell wall proteome analysis of A. (1995) Identification of the region of a 14-kilodalton
Mycobacterium smegmatis strain MC2 155. BMC Microbiol protein of Rhodococcus ruber that is responsible for the
10: 121. binding of this phasin to polyhydroxyalkanoic acid granules.
Jackson, M., Raynaud, C., Laneelle, M.A., Guilhot, C., J Bacteriol 177: 2513–2523.
Laurent-Winter, C., Ensergueix, D., et al. (1999) Inactiva- Rao, P.K., and Li, Q. (2009) Protein turnover in mycobacterial
tion of the antigen 85C gene profoundly affects the mycolate proteomics. Molecules 14: 3237–3258.
content and alters the permeability of the Mycobacterium Raschke, D., and Knorr, D. (2009) Rapid monitoring of cell
tuberculosis cell envelope. Mol Microbiol 31: 1573– size, vitality and lipid droplet development in the oleagi-
1587. nous yeast Waltomyces lipofer. J Microbiol Methods 79:
Jarlier, V., and Nikaido, H. (1994) Mycobacterial cell wall: 178–183.
structure and role in natural resistance to antibiotics. FEMS Ronning, D.R., Klabunde, T., Besra, G.S., Vissa, V.D.,
Microbiol Lett 123: 11–18. Belisle, J.T., and Sacchettini, J.C. (2000) Crystal structure
de Jong, B.C., Israelski, D.M., Corbett, E.L., and Small, P.M. of the secreted form of antigen 85C reveals potential
(2004) Clinical management of tuberculosis in the context targets for mycobacterial drugs and vaccines. Nat Struct
of HIV infection. Annu Rev Med 55: 283–301. Biol 7: 141–146.
Kalscheuer, R., and Steinbüchel, A. (2003) A novel bifunc- Ronning, D.R., Vissa, V., Besra, G.S., Belisle, J.T., and Sac-
tional wax ester synthase/acyl-CoA:diacylglycerol acyl- chettini, J.C. (2004) Mycobacterium tuberculosis antigen
transferase mediates wax ester and triacylglycerol 85A and 85C structures confirm binding orientation and
biosynthesis in Acinetobacter calcoaceticus ADP1. J Biol conserved substrate specificity. J Biol Chem 279: 36771–
Chem 278: 8075–8082. 36777. Epub 32004 Jun 36710.
Lardizabal, K.D., Mai, J.T., Wagner, N.W., Wyrick, A., Russell, D.G., Barry, C.E., 3rd, and Flynn, J.L. (2010) Tuber-
Voelker, T., and Hawkins, D.J. (2001) DGAT2 is a new culosis: what we don’t know can, and does, hurt us. Science
diacylglycerol acyltransferase gene family: purification, 328: 852–856.
cloning, and expression in insect cells of two polypeptides Saha, S., Enugutti, B., Rajakumari, S., and Rajasekharan, R.
from Mortierella ramanniana with diacylglycerol acyltrans- (2006) Cytosolic triacylglycerol biosynthetic pathway in
ferase activity. J Biol Chem 276: 38862–38869. oilseeds. Molecular cloning and expression of peanut
Lewis, K. (2007) Persister cells, dormancy and infectious cytosolic diacylglycerol acyltransferase. Plant Physiol 141:
disease. Nat Rev Microbiol 5: 48–56. 1533–1543.
Liu, Q., Siloto, R.M., and Weselake, R.J. (2010) Role Sanner, M.F. (1999) Python: a programming language for
of cysteine residues in thiol modification of acyl- software integration and development. J Mol Graph Model
CoA:diacylglycerol acyltransferase 2 from yeast. Biochem- 17: 57–61.
istry 49: 3237–3245. Satoh, T., and Hosokawa, M. (1998) The mammalian car-
Low, K.L., Shui, G., Natter, K., Yeo, W.K., Kohlwein, S.D., boxylesterases: from molecules to functions. Annu Rev
Dick, T., et al. (2010) Lipid droplet-associated proteins are Pharmacol Toxicol 38: 257–288.
involved in the biosynthesis and hydrolysis of triacylgly- Schüttelkopf, A.W., and van Aalten, D.M. (2004) PRODRG: a
cerol in Mycobacterium bovis bacillus Calmette-Guerin. tool for high-throughput crystallography of protein-ligand
J Biol Chem 285: 21662–21670. complexes. Acta Crystallogr D Biol Crystallogr 60: 1355–
McKinney, J.D., Honer zu Bentrup, K., Munoz-Elias, E.J., 1363.
Miczak, A., Chen, B., Chan, W.T., et al. (2000) Persistence Sirakova, T.D., Dubey, V.S., Deb, C., Daniel, J., Korotkova,
of Mycobacterium tuberculosis in macrophages and mice T.A., Abomoelak, B., and Kolattukudy, P.E. (2006) Identifi-
requires the glyoxylate shunt enzyme isocitrate lyase. cation of a diacylglycerol acyltransferase gene involved in
Nature 406: 735–738. accumulation of triacylglycerol in Mycobacterium tubercu-
Meena, L.S., and Rajni (2010) Survival mechanisms of losis under stress. Microbiology 152: 2717–2725.
pathogenic Mycobacterium tuberculosis H37Rv. FEBS J Stover, C.K., de la Cruz, V.F., Fuerst, T.R., Burlein, J.E.,
277: 2416–2427. Benson, L.A., Bennett, L.T., et al. (1991) New use of BCG
Murphy, D.J., and Brown, J.R. (2007) Identification of gene for recombinant vaccines. Nature 351: 456–460.
targets against dormant phase Mycobacterium tuberculo- Trott, O., and Olson, A.J. (2010) AutoDock Vina: improving
sis infections. BMC Infect Dis 7: 84. the speed and accuracy of docking with a new scoring
Nguyen, L., Chinnapapagari, S., and Thompson, C.J. (2005) function, efficient optimization, and multithreading.
FbpA-dependent biosynthesis of trehalose dimycolate is J Comput Chem 31: 455–461.
required for the intrinsic multidrug resistance, cell wall Wältermann, M., Hinz, A., Robenek, H., Troyer, D., Reichelt,
structure, and colonial morphology of Mycobacterium R., Malkus, U., et al. (2005) Mechanism of lipid-body for-
smegmatis. J Bacteriol 187: 6603–6611. mation in prokaryotes: how bacteria fatten up. Mol Micro-
Olukoshi, E.R., and Packter, N.M. (1994) Importance of biol 55: 750–763.
stored triacylglycerols in Streptomyces: possible carbon Wältermann, M., Stöveken, T., and Steinbüchel, A. (2007) Key
source for antibiotics. Microbiology 140: 931–943. enzymes for biosynthesis of neutral lipid storage com-

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592


1592 A. A. Elamin et al. 䊏

pounds in prokaryotes: properties, function and occurrence enzymes and triacylglycerol biosynthesis. J Lipid Res 49:
of wax ester synthases/acyl-CoA:diacylglycerol acyltrans- 2283–2301.
ferases. Biochimie 89: 230–242.
Wong, M.Y., Steck, P.A., and Gray, G.R. (1979) The major
mycolic acids of Mycobacterium smegmatis. Characteriza- Supporting information
tion of their homologous series. J Biol Chem 254: 5734–
Additional supporting information may be found in the online
5740.
version of this article.
Yen, C.L., Monetti, M., Burri, B.J., and Farese, R.V., Jr (2005)
The triacylglycerol synthesis enzyme DGAT1 also cata- Please note: Wiley-Blackwell are not responsible for the
lyzes the synthesis of diacylglycerols, waxes, and retinyl content or functionality of any supporting materials supplied
esters. J Lipid Res 46: 1502–1511. by the authors. Any queries (other than missing material)
Yen, C.L., Stone, S.J., Koliwad, S., Harris, C., and Farese, should be directed to the corresponding author for the
R.V., Jr (2008) Thematic review series: glycerolipids. DGAT article.

© 2011 Blackwell Publishing Ltd, Molecular Microbiology, 81, 1577–1592

You might also like