Você está na página 1de 20

Energetics of Respiration and

Oxidative Phosphorylation
in Mycobacteria
GREGORY M. COOK,1 KIEL HARDS,1 CATHERINE VILCHÈZE,2
TRAVIS HARTMAN,2 and MICHAEL BERNEY2
1
University of Otago, Department of Microbiology and Immunology, Dunedin, New Zealand; 2Department of
Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461

ABSTRACT Mycobacteria inhabit a wide range of intracellular OVERVIEW OF MYCOBACTERIAL


and extracellular environments. Many of these environments RESPIRATION AND OXIDATIVE
are highly dynamic, and therefore mycobacteria are faced with PHOSPHORYLATION
the constant challenge of redirecting their metabolic activity to
be commensurate with either replicative growth or a
The genus Mycobacterium comprises a group of obli-
nonreplicative quiescence. A fundamental feature in this gately aerobic bacteria that have adapted to inhabit a
adaptation is the ability of mycobacteria to respire, regenerate wide range of intracellular and extracellular environ-
reducing equivalents, and generate ATP via oxidative ments. A fundamental feature in this adaptation is the
phosphorylation. Mycobacteria harbor multiple primary ability to respire and generate energy from variable
dehydrogenases to fuel the electron transport chain, and sources or to sustain metabolism in the absence of
two terminal respiratory oxidases, an aa3-type cytochrome
growth. Early studies on respiration demonstrated that
c oxidase and a cytochrome bd-type menaquinol oxidase,
are present for dioxygen reduction coupled to the
Mycobacterium tuberculosis H37Rv, grown in the lungs
generation of a proton motive force (PMF). Hypoxia leads to of infected mice, had high rates of endogenous respira-
the downregulation of key respiratory complexes, but the tion that were not stimulated by exogenous substrates
molecular mechanisms regulating this expression are (e.g., acetate, pyruvate, glucose, glycerol, lactate) (1). In
unknown. Despite being obligate aerobes, mycobacteria have contrast, cells grown in vitro respired these substrates at
the ability to metabolize in the absence of oxygen, and a high rates. Fatty acids, however, stimulated the respi-
number of reductases are present to facilitate the turnover ration of in vivo–grown M. tuberculosis, suggesting for
of reducing equivalents under these conditions (e.g., nitrate
reductase, succinate dehydrogenase/fumarate reductase).
Hydrogenases and ferredoxins are also present in the genomes
Received: 26 April 2013, Accepted: 6 August 2013,
of mycobacteria, suggesting the ability of these bacteria to Published: 6 June 2014
adapt to an anaerobic type of metabolism in the absence of Editors: Graham F. Hatfull, University of Pittsburgh, Pittsburgh, PA,
oxygen. ATP synthesis by the membrane-bound F1F0-ATP and William R. Jacobs, Jr., Howard Hughes Medical Institute, Albert
synthase is essential for growing and nongrowing Einstein College of Medicine, Bronx, NY
mycobacteria, and the enzyme is able to function over a Citation: Cook GM, Hards K, Vilchèze C, Hartman T, Berney M. 2014.
wide range of PMF values (aerobic to hypoxic). The discovery Energetics of respiration and oxidative phosphorylation in
mycobacteria. Microbiol Spectrum 2(3):MGM2-0015-2013.
of lead compounds that target respiration and oxidative
doi:10.1128/microbiolspec.MGM2-0015-2013.
phosphorylation in Mycobacterium tuberculosis highlights the
Correspondence: G.M. Cook, gregory.cook@otago.ac.nz
importance of this area for the generation of new frontline
© 2014 American Society for Microbiology. All rights reserved.
drugs to combat tuberculosis.

ASMscience.org/MicrobiolSpectrum 1
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

the first time that M. tuberculosis switches to different (cydABCD) (Fig. 1). Mycobacteria coordinate the ex-
energy sources in host tissues to fuel respiration. These pression of these oxidases in response to oxygen supply
early studies pointed to the fact that electron donor to achieve maximal efficiency of oxygen utilization. The
utilization and respiration are precisely controlled in rationale for this coordinated expression is largely based
M. tuberculosis, not only in response to growth rate, on the assumption that the mycobacterial aa3-type cyto-
but also in response to the carbon and energy sources chrome c oxidase and cytochrome bd-type have markedly
used for growth. The pioneering work of Brodie and different affinities for oxygen in mycobacteria, but this
colleagues on Mycobacterium phlei established much of remains to be experimentally shown. Furthermore, the
the primary information on the electron transport chain molecular mechanisms governing the regulation of ter-
and oxidative phosphorylation system in mycobacteria minal oxidase expression in mycobacterial species are
(reviewed in reference 2). unknown. In the absence of oxygen, mycobacterial
Since these early studies, comparatively few biochemi- growth is inhibited, even if alternative electron acceptors
cal studies have been performed on the electron trans- are present (e.g., nitrate, fumarate). Despite growth being
port components and the energetics of respiration in inhibited, mycobacteria are able to metabolize exogenous
mycobacterial species. With the advent of microbial ge- and endogenous energy sources under low oxygen for
nome sequencing, sequence analyses have revealed that maintenance functions. Whether mycobacteria utilize
branched pathways exist in mycobacterial species for energy spilling or overflow metabolism (3) under these
electron transfer from many low potential reductants via conditions to balance the rate of catabolic versus ana-
quinol, including H2, to oxygen (Fig. 1). Unlike other bolic reactions remains to be determined. The electron
bacteria, there appears to be little redundancy in the acceptors and mechanisms to recycle reducing equivalents
transfer of electrons to oxygen during growth with only under these conditions are poorly understood. Irrespective
two terminal respiratory oxidases present in myco- of the oxygen concentration or the proton motive force
bacteria, an aa3-type cytochrome c oxidase (encoded by (PMF), ATP synthesis is obligatorily coupled to the elec-
ctaBCDE) belonging to the heme-copper respiratory ox- tron transport chain and the F1F0-ATP synthase, but the
idase family and cytochrome bd-type menaquinol oxidase reasons for this remain unexplained.

FIGURE 1 Organization and components of the electron transport chain in mycobacteria.


doi:10.1128/microbiolspec.MGM2-0015-2013.f1

2 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

Research into mycobacterial respiration and oxida- and valinomycin), and these compounds are bactericidal
tive phosphorylation has been energized by the discov- toward growing and nongrowing (aerobic or hypoxic)
ery of a new drug (TMC207; Sirturo; Bedaquiline) that cells, further highlighting the importance of the mem-
targets the ATP synthase of mycobacteria, suggesting brane potential in mycobacterial viability (4, 6). Growth
that inhibitors of respiration and ATP synthesis will is also sensitive to the ionophore monensin, but the
provide the next generation of front-line drugs to com- reasons for this are not clear. Interestingly, some solute
bat tuberculosis and nontuberculous mycobacterial dis- transporters in mycobacteria are driven by a sodium-
ease. The aim of this article is to discuss recent advances motive force (7), but the mechanism used for the gen-
in understanding the processes of respiration and eration of the primary sodium gradient in mycobacteria
oxidative phosphorylation in mycobacteria, with a is not known. As the pH of the growth medium changes
particular focus on the obligate human pathogen and becomes mildly acidic, mycobacteria are able to
M. tuberculosis. generate a considerable transmembrane pH gradient
(ZΔpH) and maintain a constant PMF (4). While proton
translocation via the respiratory chain generates the
PMF during respiration with oxygen as the terminal
ENERGETICS OF MYCOBACTERIA DURING electron acceptor, it is not clear how the PMF is
GROWTH AND NONGROWTH CONDITIONS established in the absence of oxygen under anaerobic
Mycobacteria generally grow at neutral pH and un- growth conditions. Anaerobic bacteria are able to gen-
der these conditions generate a PMF of approximately erate a significant PMF (−100 mV) using their mem-
−180 mV (4). The PMF generated is used to drive proton- brane-bound F1F0-ATP synthase in the ATP hydrolysis
coupled energetic processes (e.g., ATP synthesis, solute direction (8). The ATPase activity (proton pumping) of
transport, etc.) (4). The PMF-generating machinery in the enzyme is fueled by ATP produced by substrate-level
mycobacteria comprises three energy-conserving steps: phosphorylation. This mechanism does not appear to
complex I (4H+/2e−), complex III (menaquinol-cytochrome operate in mycobacterial cells, where the F1F0-ATP
c oxidoreductase, 4H+/2e−), and complex IV (2H+/2e−), synthase has been reported to have latent ATPase ac-
suggesting that the overall proton translocation stoichi- tivity when measured in inverted membrane vesicles (9,
ometry for the transfer of 2e– from NADH to oxygen is 10). Whether the enzyme is also latent in actively
10H+/2e−. A ratio of 3H+ utilized by the ATP synthase/ growing cells is not known, and therefore the potential
ATP synthesized leads to a theoretical maximum P/O exists for this enzyme to function as a primary proton
ratio (i.e., the number of moles of ADP phosphorylated to pump in the absence of oxygen and a functional respi-
ATP per 2e− passing to oxygen) of approximately 3.3 ratory chain to generate the PMF. Rao et al. (6) have
(theoretical maximum). If complex I is bypassed by the reported that hypoxic, nonreplicating M. tuberculosis
non-proton translocating type II NADH:menaquinone generates a total PMF of −113 mV, −73 mV of electrical
oxidoreductase, the P/O ratio would be approximately potential (ΔΨ), and −41 mV of ZΔpH. The addition of
2. Electron flow from the menaquinone pool to the thioridazine, a compound that targets NDH-2, results
cytochrome bd oxidase branch (bypassing complex III in dissipation of the ΔΨ and significant cell death,
and IV) would produce a P/O ratio of 0.67. Measured suggesting that NADH is an important electron donor
experimental values for mycobacteria oxidizing NADH for the generation of the ΔΨ under hypoxic conditions.
or succinate yield P/O ratios of 0.52 and 0.36, respec- The addition of TMC207, a specific inhibitor of the
tively (5). Variations in theoretical P/O ratios versus F1F0-ATP synthase, was bactericidal against hypoxic,
those determined experimentally are well accepted and nonreplicating M. tuberculosis but had no effect on the
can be explained by pathways that involve proton ΔΨ (6).
leakage (i.e., bypass ATP synthase), or the PMF is used
to drive reverse electron transport. When succinate
oxidation is coupled to menaquinone reduction, the
energetics suggest a reverse electron flow from succinate ELECTRON DONORS FUELING RESPIRATION
(lower midpoint redox potential Em = +30 mV) to IN MYCOBACTERIA
menaquinone (Em = −74 mV), resulting in lower P/O Mycobacterial species use a variety of primary de-
ratios. hydrogenases to deliver electrons from central metab-
Growth of mycobacteria is sensitive to compounds that olism into the respiratory chain to generate energy
dissipate the membrane potential (e.g., protonophores (Fig. 1).

ASMscience.org/MicrobiolSpectrum 3
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

Proton-Pumping Type I NADH Dehydrogenase lower than the NDH-2 activity when measured in mem-
and Non-Proton-Pumping Type II brane fractions of mycobacteria (C. Vilchèze and W. R.
NADH Dehydrogenase Jacobs, Jr., unpublished data), suggesting that the major
The major entry point is the transfer of electrons from NADH oxidizing activity is mediated by NDH-2. This is
NADH oxidation to quinone reduction (e.g., ubiqui- supported by the observation that NADH oxidation by
none or menaquinone). In bacteria, three different mycobacterial membranes is relatively insensitive to
types of respiratory NADH dehydrogenases have been complex I inhibitors (12). Notwithstanding this, M. tu-
identified and characterized on the basis of reaction berculosis mutants lacking only one of the subunits of
mechanism, subunit composition, and protein architec- NDH-1, nuoG, have an in vivo phenotype, where this
ture (11). These include the proton-pumping type I gene was critical for host macrophage apoptosis inhibi-
NADH dehydrogenase (NDH-1, complex I), non- tion and mouse virulence (17). This implies that
proton-pumping type II NADH dehydrogenase (NDH- nuoG and potentially other subunits of NDH-1 are anti-
2), and sodium-pumping NADH dehydrogenase (NQR). apoptosis factors and are attractive candidates for
Weinstein et al. identified genes for two classes of NADH: vaccine development.
menaquinone oxidoreductases in the genome of M. Several studies have reported that nuo is down-
tuberculosis (12) (Table 1). NDH-1 is encoded by the regulated in M. tuberculosis during mouse lung infection
nuoABCDEFGHIJKLMN operon and transfers elect- (18), during survival in macrophages (19), in both
rons to menaquinone, conserving energy by translocating nonreplicating persistence (NRP)-1 (1% oxygen satu-
protons across the membrane to generate a PMF (Fig. 2). ration) and NRP-2 (0.06% oxygen saturation) relative
The second class is NDH-2, a non-proton-translocating to aerated mid-log growth (18), and upon starvation
type II NADH dehydrogenase that does not conserve en- in vitro (20). The transcription of NDH-2(ndh) is also
ergy and is present in two copies (Ndh and NdhA) in M. downregulated in M. tuberculosis during mouse lung
tuberculosis (12) (Table 1). Sodium-pumping NADH de- infection, but transcript levels for ndh peak during
hydrogenase has not been reported in mycobacterial NRP-2 in vitro, demonstrating that the pattern of ndh
genomes. regulation is different between in vivo and in vitro
NDH-1 is composed of 14 subunits (nuoA-N), which conditions (19). These data are in contrast to Esche-
represent a 15,704-bp operon in M. tuberculosis (Rv3145 richia coli, in which NDH-1 is usually associated with
to Rv3158) (Table 1). NuoB, C, D, E, F, and G are pe- anaerobic respiratory pathways (e.g., fumarate) and
ripheral membrane proteins located in the cytoplasmic noncoupling dehydrogenases such as NDH-2 are syn-
side, while NuoA, H, J, K, L, M, and N are in the thesized aerobically (21).
membrane section of the complex with multiple predicted The non-proton-translocating NDH-2 is a small mono-
transmembrane domains (from 3 to 16). In contrast, the topic membrane protein (50 to 60 kDa) that catalyzes
nuo operon has been lost from the genome of the intra- electron transfer from NADH via FAD (noncovalently
cellular parasite Mycobacterium leprae except for a single bound redox prosthetic group) to quinone. No tertiary
remaining nuoN pseudogene (13). NDH-1 uses flavin structural information exists for either the bacterial,
mononucleotide (FMN) and iron-sulfur clusters to plant, or protist NDH-2 enzymes, but the yeast NDH-2
transport electrons from NADH to the quinone pool structure was recently solved by two laboratories (22,
(menaquinone). The release of the two electrons during 23). NDH-2 is widespread in bacteria and the mito-
the NADH oxidation produces enough energy to pump chondria of fungi, plants, and some protists. In some
four protons across the membrane to generate a PMF cases more than one copy is present (24). The role(s) of
(Fig. 2). In M. tuberculosis, the nuo operon is essential multiple type II NADH dehydrogenases in prokaryotes,
for neither growth nor persistence in an in vitro Wayne plants, and parasites is unclear. In some organisms with
model (6). Mycobacterium smegmatis also contains multiple type II NADH dehydrogenases, one copy
genes for a type I NADH:menaquinone oxidoreductase appears more essential than the other, pointing to non-
(nuoA-N). However, M. smegmatis NDH-I activity is redundant functional differences (25, 26). M. tubercu-
very low, representing about 5% of the NDH-2 activity losis harbors two copies of NDH-2 (ndhRv1854c and
(14, 15). Rather, increased expression (15-fold) of the ndhA Rv0392c) (12) (Table 1), which are well conserved
nuo operon in M. smegmatis was observed in a carbon- among slow-growing mycobacterial species. In M. tu-
limited chemostat in response to a slowdown in growth berculosis, Ndh (1,392 bp) and NdhA (1,413 bp) share
rate (16). In the slow-growing M. tuberculosis and My- 65% identity; however, the FAD and NADH binding
cobacterium bovis strains, NDH-1 activity is 28 to 50% motifs, G21SGFGG26 and G177AGPTG182, are highly

4 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

conserved. Both proteins contain one transmembrane may represent a potential drug target for the treatment
domain located at the amino acids 385 to 407 and 387 of human pathogens (6, 12, 27, 30, 31) and intracellular
to 409, respectively. The Ndh and NdhA proteins of M. parasites (32). Despite the potential of NDH-2 as a drug
tuberculosis have been shown to be functional NADH target, no potent nanomolar inhibitors of NDH-2 are
dehydrogenases that transfer electrons to the quinone known. Several classes of compounds are proposed to
pool via a ping-pong reaction mechanism (27). NdhA is target the enzyme at micromolar concentrations (e.g.,
not present in M. smegmatis, yet the level of NADH phenothiazine analogues, platanetin, quinolinyl pyri-
oxidation by M. smegmatis NDH-2 is several-fold midines) (12, 27, 33), but the mechanism of inhibition
higher than in M. tuberculosis or M. bovis and remains unknown. Despite poor in vitro activity, drugs
represents 95% of the total NADH oxidation measured of the phenothiazine family (trifluoroperazine, chlor-
(15). This might correlate with higher NAD+ con- promazine) have potent activity in vitro against drug-
centrations in M. smegmatis compared to M. bovis susceptible and drug-resistant M. tuberculosis strains
(three times higher, as shown in reference 15). In addi- (34, 35). A phenothiazine analog was also tested in a
tion, when M. smegmatis and M. bovis were trans- mouse model of acute M. tuberculosis infection and was
formed with a replicative plasmid containing ndh, the found to reduce by 90% the M. tuberculosis bacterial
ratio NAD+/NADH doubled (15), further confirming load in the lungs after 11 days of treatment compared to
the involvement of ndh in the oxidation of NADH a 3- to 4-log reduction in CFUs with the INH or rifampin
into NAD+. control (12). From a library of microbial products, two
Several studies have suggested that ndh is essential new compounds, scopafungin and gramicidin S, were
for growth of M. tuberculosis (12, 26, 28, 29) (Table 1). identified as inhibitors of M. smegmatis NDH-2, with
Mutations in the ndh gene of M. smegmatis result in IC50 values better than trifluoperazine (36). There is a
a pleiotropic effect: temperature sensitivity, amino acid crucial need for new drug targets to inhibit M. tuber-
auxotrophy, and resistance to the first-line anti-TB drug culosis, and the electron transport chain is a very at-
isoniazid (INH) and its analog and second-line anti-TB tractive avenue. However, reduction in NDH-2 activity
drug ethionamide (ETH) (14, 15). The ndh mutants had has been linked to INH and ETH resistance in both
decreased NADH oxidase activity and increased NADH slow- and fast-growing mycobacteria (15), and pheno-
concentration (14, 15). Selection for ndh mutants grown thiazines have been shown to be antagonistic with INH
on rich media (Mueller Hinton) led to the isolation of (30). Therefore, the development of NDH-2 inhibitors
ndh mutants that were auxotrophic for serine and gly- will have to ascertain that interference with current drug
cine; this auxotrophy was resolved with complementa- therapy does not occur.
tion by a wild-type copy of ndh. The correlation between Some interesting questions arise from these obser-
ndh mutations and serine/glycine auxotrophy was at- vations. Why do mycobacteria use type II NADH
tributed to the increase in NADH concentration, which dehydrogenases to recycle NADH, when they could
might inhibit the first step in serine/glycine biosynthesis continue to use the energy-conserving and PMF-
(14). The increase in NADH concentration was also generating NDH-I? One potential explanation is that
responsible for the high resistance to INH and ETH by because type II NADH dehydrogenases are non-proton-
competitively inhibiting the binding of the INH-NAD or translocating, they will not be impeded by a high PMF,
ETH-NAD adduct to the NADH-dependent enoyl-ACP which could ultimately slow down metabolic flux due to
reductase InhA (15). ndh mutants in both slow-growing back-pressure on the system. This mechanism is akin to a
(M. bovis BCG) and fast-growing (M. smegmatis) my- “relief valve” that would allow for a higher metabolic
cobacteria had no growth defect at permissive temper- flux and ultimately higher rates of ATP synthesis, at the
ature, although they lost up to 90% of their ability to expense of low energetic efficiency of the respiratory
oxidize NADH. Interestingly, in M. smegmatis and chain. Second, why is ndh an essential gene when
M. bovis, the levels of NAD+ cofactor stayed relatively mycobacteria could also use nuo? The fact that ndh is
constant despite overexpression of ndh or mutations in essential implies that mycobacteria do not have another
ndh, which highly reduced their NADH oxidation ca- mechanism to recycle NADH during normal aerobic
pability (15), suggesting that the NAD+ pool is tightly growth. Alternatively, this is the only NADH dehydro-
regulated in mycobacteria to maintain essential bio- genase that operates under these growth conditions,
chemical functions. and the activity of this enzyme is essential for main-
NDH-2 has not been reported in mammalian mito- taining an energized membrane. Compounds that target
chondria, leading to the proposal that these enzymes NDH-2 are bactericidal toward hypoxic nonreplicating

ASMscience.org/MicrobiolSpectrum 5
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
6
Cook et al.

TABLE 1 Electron transport chain components and energy-generating machinery of mycobacteria

In vitro essentiality

Operon/genea Subunits Rv # Enzyme name Griffin et al. (26) Zhang et al. (129)b
nuo nuoABCDEFGHIJKLMN Rv3145–Rv3158 Type I NADH:menaquinone nuoDFGHI = yes nuoDFGL = yes
oxidoreductase
ndh Rv1854c Type II NADH:menaquinone Yes No
oxidoreductase
ndhA Rv0392c Type II NADH:menaquinone No No
oxidoreductase
sdh1 sdh1CD Rv0249c Succinate:menaquinone Yes Only sdh1A
sdh1A Rv0248c oxidoreductase I
sdh1B Rv0247c
sdh2 sdh2C Rv3316 Succinate:menaquinone No No
sdh2D Rv3317 oxidoreductase II
sdh2A Rv3318
sdh2B Rv3319
mqo Rv2852c Malate:menaquinone No No
oxidoreductase
pru pruA Rv1187 Proline dehydrogenase and Yes Yes
pruB Rv1188 pyrroline-5-carboxylate

IP: 129.82.52.133
dehydrogenase
gpdA1 Rv0564c Glycerol-3-phosphase No No

On: Sat, 17 Mar 2018 19:24:45


dehydrogenase A1
gpdA2 Rv2982c Glycerol-3-phosphase No No

Downloaded from www.asmscience.org by


dehydrogenase A2
glpD1 Rv2249c Glycerol-3-phosphase No Yes
(Rv2250c) dehydrogenase D1
glpD2 glpD2 Rv3302c Glycerol-3-phosphase Yes Yes
ldpA Rv3303c dehydrogenase D2
Dihydrolipoamide
dehydrogenase
cox coxCMSLDEFG Rv0376c–Rv0368c Carbon monoxide coxLEF = Yes coxCL = yes
dehydrogenase
ald Rv2780 L-Alanine dehydrogenase No No
lldD1 ? Rv0692 L-Lactate dehydrogenase 1 No lldD1 = yes
pqqE Rv0693
lldD1 Rv0694
lldD2 lldD2 Rv1872c L-Lactate dehydrogenase 2 No No
Rv1871c
Rv1870c
Rv0843 (Rv0842) Pyruvate dehydrogenase No No

ASMscience.org/MicrobiolSpectrum
Rv0843 E1 component alpha subunit
pdh pdhA Rv2497c Pyruvate dehydrogenase pdhB = yes pdhC = yes
pdhB Rv2496c
pdhC Rv2495c
ace Rv2241 Pyruvate dehydrogenase Yes Yes
E1 component
lpd Rv0462 Dihydrolipoamide Yes Yes
dehydrogenase
hyc Rv0081 Rv0081-Rv0088 Formate hydrogenlyase OR hycE and hycQE = yes
Rv0082 energy-converting Rv0088 = yes
Rv0083 hydrogenase-related
hycDPQE complex (Ehr) (a.k.a HydTB)
Rv0088
qcr qcrC Rv2194 Cytochrome bc1 Yes Yes
qcrA Rv2195
qcrB Rv2196
ctaB Rv1451 Cytochrome c oxidase Yes Yes

ASMscience.org/MicrobiolSpectrum
assembly factor
ctaC ctaC Rv2200c Transmembrane cytochrome Yes Rv2200c = Yes
(Rv2199c) c subunit II
ctaD Rv3043c Cytochrome c oxidase Yes Yes
polypeptide I
ctaE Rv2193 Cytochrome c oxidase Yes Yes
subunit III
cyd cydA Rv1623c Cytochrome bd oxidase Yes cydBC = yes
cydB Rv1622c
cydD Rv1621c
cydC Rv1620c
nar narG Rv1161 Menaquinone:nitrate No No

IP: 129.82.52.133
narH Rv1162 reductase
narJ Rv1163

On: Sat, 17 Mar 2018 19:24:45


narI Rv1164
sirA sirA Rv2391 Sulfite reductase Yes Rv2391 and

Downloaded from www.asmscience.org by


cysH Rv2392 APS reductase Rv2392 = yes
che1 Rv2393 Ferrochelatase
nirBD nirB Rv0252 Nitrite reductase [NAD(P)H] No No
nirD Rv0253
frd frdA Rv1552 Menaquinone:fumarate No frdA = yes
frdB Rv1553 reductase
frdC Rv1554
frdD Rv1555
atp atpIBEFHAGDC and Rv1312 Rv1303-Rv1312 F1F0 ATP synthase operon Yes Yes except atpI
and Rv1312

aIn the absence of operons confirmed by previous publications, the TB Database operon prediction algorithms (www.tbdb.org) were used to identify potential operons. Rv loci enclosed in parentheses signify weak

operon predictions.
bN.B. Genes identified as containing both essential and nonessential regions (a “D call”) are referred to as essential here.
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

7
Cook et al.

FIGURE 2 The core respiratory chain of mycobacteria and components upregulated


under energy-limiting conditions. During in vitro exponential growth, mycobacteria
use a classical respiratory chain composed of a type I NADH:menaquinone oxido-
reductase (Nuo), succinate:menaquinone oxidoreductase 1 (SDH1), cytochrome aa3-bc
supercomplex (Qcr-Cta), and F1F0 ATPase. Menaquinone (MQ) is the only quinone present
in mycobacterial membranes, and reverse electron transport driven by the PMF is pro-
posed to facilitate the function of SDH1 and similar enzymes (see text). Components in
light blue are upregulated in response to energy-limiting conditions (6). Catalysis and
electron flow are indicated by arrows. Abbreviations: Cox, carbon monoxide dehydro-
genase; Hyd, hydrogenase; DH, dehydrogenase; A, unidentified electron acceptor.
doi:10.1128/microbiolspec.MGM2-0015-2013.f2

M. tuberculosis, suggesting that the respiratory chain is catalyzes the oxidation of succinate to fumarate wherein
essential for the recycling of NADH under these con- two electrons are transferred to quinol. The reverse re-
ditions (6). action can be catalyzed by fumarate reductase (FRD),
which is involved in anaerobic respiration (Fig. 3).
FRD and succinate dehydrogenase are closely related
Multiple Succinate Dehydrogenases and enzymes, and the reaction catalyzed cannot be predicted
Fumarate Reductase: An Essential Link based on the primary sequence alone. Most mycobac-
Between Central Metabolism and Respiration teria harbor two annotated succinate dehydrogenases,
in Mycobacteria SDH1 and SDH2 (Table 1). SDH2 has high homology
Succinate dehydrogenase forms complex II of the respi- to SDH enzymes from other species and is encoded by
ratory chain and couples oxidative phosphorylation to four genes, sdhC, sdhD, sdhA, and sdhB. The genes
central carbon metabolism by being an integral part of sdhA and sdhB encode for the cytoplasmic part of the
the tricarboxylic acid (TCA) cycle (Fig. 2). This enzyme enzyme where the succinate to fumarate reaction takes

8 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

FIGURE 3 The preferential respiratory chain of an oxygen-limited mycobacterial cell.


Under low-oxygen conditions, a diverse response utilizing alternate electron donors and
acceptors, energy-conserving enzymes, and a high-affinity terminal oxidase permits
survival under hypoxic conditions. Components in red are upregulated under microaerobic
conditions (6). Catalysis and electron flow are indicated by arrows. The possible PMF-
driven reverse electron flow of Sdh2 is not shown, for clarity. Abbreviations: Mqo, malate:
menaquinone oxidoreductase; Ndh, type II NADH:menaquinone oxidoreductase; Sdh2,
succinate:menaquinone oxidoreductase 2; Nar, nitrate reductase; Cyd, cytochrome bd
oxidase; Frd, FRD; Hyd, hydrogenase; MQ, menaquinone; A, unidentified electron ac-
ceptor. doi:10.1128/microbiolspec.MGM2-0015-2013.f3

place (SdhA), and electrons are shuttled via three iron- mycobacterial species (37, 38), but it is still unclear
sulfur clusters (SdhB) to the membrane subunits SdhC which of the three enzymes, or all, are responsible for
and SdhD, which catalyze the electron transfer to me- this activity.
naquinone. The SdhA (encoded by Rv0248c) and SdhB Mycobacteria utilize menaquinone/menaquinol (MQ/
(Rv0247c) subunits of SDH1 are similar to the common MQH2) as a conduit between electron-donating and
SDH enzyme. However, the other two genes (Rv0250c -accepting reactions (Fig. 1). Menaquinone has a lower
and Rv0249c) in the operon show no homology to midpoint redox potential (Em = −74 mV) compared to
membrane-bound components of SDH and FRD and ubiquinone (Em = +113 mV) and is ideally poised to
are specific to the phylum Actinobacteria. Gene ex- donate electrons to fumarate during anaerobic con-
pression data show that all four genes of SDH1 in ditions (39). This means that the SDH reaction in
M. smegmatis are expressed in concert (16). As an ex- mycobacteria (succinate oxidation to fumarate) should
ception among mycobacteria, M. tuberculosis encodes a have an unfavorable free energy due to reverse electron
third complex (frdABCD) that is annotated as an flow and proton uptake to drive this reaction. It is
FRD (Table 1) and is absent in all other pathogenic tempting to propose that the unusual subunits of SDH1
strains such as Mycobacterium avium paratuberculosis, could be the result of structural specializations in the
Mycobacterium marinum, Mycobacterium ulcerans, and transmembrane region to facilitate reverse electron flow
M. leprae. Succinate dehydrogenase activity has been from succinate to menaquinone. In fact, SDH1 and
measured in M. tuberculosis as well as many other SDH2 have been shown to be differentially expressed

ASMscience.org/MicrobiolSpectrum 9
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

under energy- or oxygen-limiting conditions in M. The role of the frdABCD operon in the M. tubercu-
smegmatis (16). Under energy-limiting conditions SDH1 losis complex is not clear. Increased expression of this
was upregulated 4-fold, while SDH2 was downregulated operon has been shown during carbon starvation (20),
3-fold. Under oxygen-limiting conditions, SDH1 was oxygen depletion (44), and in macrophages (19), sug-
downregulated 30-fold, while SDH2 was upregulated gesting a role for this enzyme in persistence.
2-fold. This indicates that SDH1 could be the dedi-
cated succinate dehydrogenase, and SDH2 catalyzes
FRD activity, which is important for survival under Alternative Electron Donor Utilization During
hypoxia. This hypothesis is supported by several reports Starvation and Hypoxia
in the recent literature. A transposon-site hybridization During carbon starvation and slow growth, myco-
(TraSH) screen in M. tuberculosis suggested that SDH1 bacteria switch to alternative electron donors (16, 20, 45)
but not SDH2 was essential for growth under aerobic (Fig. 2). Proline dehydrogenase is upregulated under
conditions on standard medium (26). In contrast, in a both energy-limiting conditions and hypoxia (7, 16) and
TraSH screen selecting for mutants that continue to is increasingly being recognized as a critical amino acid in
replicate under hypoxic conditions, Baek and coworkers cellular bioenergetics and redox control (46). Proline can
(40) found SDH2 mutants overrepresented. These be utilized as an electron donor as well as a carbon and
results suggest that SDH2 has a pivotal role in the nitrogen source (Fig. 2). The degradation of proline
transition of M. tuberculosis from aerobic to hypoxic occurs by means of two enzymes: proline dehydrogenase
conditions and supports the hypothesis of its being an (PRODH) and pyrroline-5-carboxylate dehydrogenase
FRD. (P5CDH). These two enzymes catalyze the oxidation of
In fact, it has been proposed that fumarate may be an proline to glutamate with four electrons transferred to
important endogenous electron acceptor for energy the respiratory chain (46). In the first step FAD is reduced
production and maintenance of redox balance (oxidation to FADH2, while in the second step NAD+ is reduced to
of NADH to NAD+) in hypoxic nonreplicating myco- NADH. In some bacteria, PRODH and P5CDH are
bacteria (6). Interestingly, the use of fumarate as an monofunctional enzymes, but in the majority of bacterial
electron acceptor in E. coli requires complex I, and ex- species they are fused into one protein called proline
pression of the nuo operon is stimulated by the presence utilization A flavoenzyme PutA (47).
of fumarate (41). This stands in direct contrast to In mycobacteria, PRODH and P5CDH are predicted
M. tuberculosis, where the nuo operon seems to be silent to be monofunctional enzymes (46). The genes encoding
under anaerobic conditions (18). In a recent study it was PRODH (putB, Rv1188) and P5CDH (putA, rocA,
shown by 13C flux analysis that M. tuberculosis grown Rv1187) are expressed as part of an operon (7). It has
under hypoxia metabolizes glucose through a reverse been shown that M. smegmatis can grow on proline as
TCA cycle to generate succinate as an excreted fermen- the sole carbon and energy source and also that PRODH
tation end product (42). However, the metabolic flux is an important electron donor, under both energy-
from fumarate to succinate was unchanged in an M. tu- limiting conditions and hypoxia (7, 16). The same
berculosis FRD deletion mutant, suggesting that one of authors showed that proline metabolism in mycobacteria
the remaining putative succinate dehydrogenases (most is regulated by a unique membrane-associated tran-
likely SDH2) could catalyze this reaction. A more recent scriptional regulator called PruC (Rv1186c), encoded
study suggests that the glyoxylate shunt, and not the re- upstream of pruA. The pruAB operon with its upstream
verse TCA, is used to metabolize both glycolytic and fatty regulator pruC is highly conserved in mycobacteria, with
acid carbon sources in response to oxygen limitation, and the exception of M. leprae. Recent proteomics data on M.
this route also produces succinate as its metabolic end avium paratuberculosis show that protein levels of RocA
product (43). The authors propose that during oxygen (PutA) are elevated in the intestinal tissues of cows (48).
limitation large amounts of succinate are produced by The authors propose that M. avium paratuberculosis has
this pathway that are used to sustain the membrane po- adapted to utilize proline as a carbon and nitrogen source
tential, ATP synthesis, and TCA cycle precursors akin due to the high abundance of this amino acid in the plant
to a “metabolic battery” (43). Moreover, because of material that is eaten by such ruminants. Mycobacteria
the near neutral midpoint potential of the succinate/ also encode pyrroline-5-carboxylate reductase (encoded
fumarate redox couple (+30 mV), succinate is able to by proC) that catalyzes the reverse reaction of putA,
bridge both oxidative and fermentative metabolic schemes converting pyrroline-5-carboxylate to proline. Interest-
depending on electron acceptor availability (43). ingly, a proC mutant of M. tuberculosis was avirulent in

10 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

immunocompetent mice, but this was not due to its in- oxidation of CO to CO2 by the following reaction: CO +
ability to proliferate intracellulary because bacterial H2O → CO2 + 2H+ + 2e−. Several pathogenic and non-
loads increased in the mouse lungs after 20 days postin- pathogenic mycobacteria including M. tuberculosis are
fection (49). known to possess CO-DH genes. It has been shown that
Hydrogenases catalyze the reversible oxidation of M. tuberculosis H37Ra, which possesses CO-DH activity,
molecular hydrogen: 2H+ + 2e− → H2 and play a central can grow on CO as a sole source of carbon and fuel for
role in energy metabolism of bacteria, archaea, and energy generation (56) (Fig. 2).
eukarya (50). Under physiological conditions, hydro- Glycerol-3-phosphate dehydrogenase catalyzes the
genases couple H2 oxidation to respiration (Knallgas oxidation of glycerol-3-phosphate to dihydroxy-acetone
reaction) or reduce protons as a way to dispose of surplus phosphate and reduces either quinone or NADP (57). In
reducing equivalents. Four different types of hydro- E. coli, glycerol-3-phosphate is used either as a precursor
genases are found in mycobacteria and are annotated to in the biosynthesis of phospholipids or as a carbon
be of the NiFe type. M. smegmatis harbors three (desig- source for energy supply (58). M. tuberculosis possesses
nated Hyd1, 2, and 3) of the four hydrogenase com- genes for four predicted glycerol-3-phosphate dehydro-
plexes (16). Hyd1 aligns closely with group 2a uptake genases, but their role and function in mycobacterial
hydrogenases of the cyanobacteria such as Nostoc, in- respiration remain unknown (Table 1).
dicating that it oxidizes H2 (51). In contrast, Hyd3 is The membrane-associated malate quinone oxidore-
closely related to the group 3 cytoplasmic bidirectional ductase (MQO) oxidizes malate to oxaloacetate and
hydrogenases. Hyd2 appears to be a founding member of transfers the reducing equivalents to menaquinone (59,
the group 5 high-affinity hydrogenases (52, 53). Hyd1, 60) (Fig. 3). M. tuberculosis harbors a copy of MQO and
Hyd2, and Hyd3 are all soluble hydrogenases and are a cytoplasmic NAD+-dependent malate dehydrogenase
found in mycobacteria of the slow-growing and fast- (MDH) (61, 62). The function and role of MQO in
growing type, as well as pathogenic and nonpathogenic mycobacterial respiration is unknown. M. smegmatis
mycobacteria (16). However, the fourth putative hy- lacks an MDH homolog, and it was shown that MQO
drogenase is only found in pathogenic mycobacteria responded to low oxygen concentration with a 4-fold
(including M. tuberculosis complex) and seems to be increase in gene expression (16). Mutants of Coryne-
restricted to slow growers (Table 1). It shows homology bacterium glutamicum defective in NDH-2 activity are
to group 4 membrane-bound H2 evolving hydrogenases. able to grow despite the loss of all membrane-bound
It has been shown that M. smegmatis, among other my- NADH dehydrogenase activity (59). The authors pro-
cobacterial species, can oxidize molecular hydrogen in pose a reaction scheme whereby electron transfer from
the presence of carbon monoxide (CO), implying that NADH to menaquinone is catalyzed by the sequential
M. smegmatis expresses a functional hydrogenase (54). action of MDH and MQO (membrane-bound). MDH
To date, no studies have reported on the ability of can reduce oxaloacetate with NADH to malate, and then
mycobacteria to produce hydrogen. Gene expression malate is reoxidized to oxaloacetate by MQO using
studies suggest that Hyd1 and Hyd2 are used during menaquinone as an electron acceptor (59, 60). Support
nutrient starvation as an alternative electron source for this model comes from the observation that a
(Fig. 2), while Hyd3 and the membrane-bound hydrog- Δmqo/ndh double mutant failed to grow under
enase are more likely to have a function in disposing of conditions where the Δndh mutant grew. Furthermore,
electrons under anaerobic conditions (16) (Fig. 3). A M. smegmatis ndh mutants could be complemented by
knockout mutant of Hyd2 in M. smegmatis showed re- M. bovis BCG mdh, suggesting that such a reaction
duced biomass production when grown on complex scheme might operate in mycobacteria (15).
medium under atmospheric conditions (16), and its ho-
molog in Streptomyces sp. was shown to facilitate hy-
drogen oxidation (53). These data suggest that Hyd2 TERMINAL ELECTRON
oxidizes hydrogen at very low concentrations, which fits ACCEPTOR UTILIZATION
with its purported role as a high-affinity hydrogenase During aerobic respiration, energy is conserved by the
(52). generation of a PMF across a proton-impermeable
Carbon monoxide dehydrogenase (CO-DH) is respon- membrane. The electron transport chain components are
sible for the oxidation of CO to carbon dioxide (CO2) in membrane bound and asymmetrically arranged across
carboxydobacteria, which grow on CO as a sole source of the membrane to achieve net consumption of protons
carbon and energy (55). Carboxydobacteria catalyze the from the cytoplasm and net release of protons on the

ASMscience.org/MicrobiolSpectrum 11
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

outside of the cell. An important part of all electron qcrCAB operon, complex III) and an aa3-type cyto-
transport chains is the terminal respiratory oxidases. In chrome c oxidase (encoded by the genes ctaBCDE,
order for mycobacteria to utilize oxygen efficiently and complex IV) belonging to the heme-copper respiratory
obtain the maximum growth yield on a particular carbon oxidase family (69–71) (Table 1). The bc1 complex
and energy source, there must be coordinate regulation (menaquinol-cytochrome c oxidoreductase) consists of
of terminal respiratory oxidase expression/activity. For redox groups comprising a 2Fe/2S center, located on a
example, in E. coli, cytochrome bo (Km for oxygen in the Rieske protein (QcrA); two b-type hemes (low and
micromolar range) and cytochrome bd (Km for oxygen in high potential) located on a single polypeptide (QcrB);
the nanomolar range) (63, 64) are coordinately regulated and the heme of cytochrome c1 (QcrC). For every two
by the ArcBA system and transcriptional regulator FNR electrons passing from quinol to cytochrome c, complex
(65). Cytochrome bo is synthesized at high oxygen ten- III releases four protons into the periplasmic side of the
sion (optimal from 15 to 100% air saturation) and re- membrane (4H+/2e−) (Fig. 2). The iron sulfur subunit
pressed as the oxygen concentration decreases (66). This QcrA has three transmembrane helices and sequence
coincides with the induction of cytochrome bd at motifs characteristic of 2Fe-2S Rieske iron-sulfur pro-
7% air saturation, which is turned off (FNR-mediated teins (e.g., CSHLGC and CPCH). In contrast to bovine
repression) once the cells enter anaerobiosis (65, 66). heart cytochrome b, QcrB from M. tuberculosis has a
Mycobacteria adopt regulation of oxidase expression to 120-amino-acid extension at the C-terminus as noted
match oxygen supply. Under conditions of low oxygen for C. glutamicum and Streptomyces species (72). The
tension (ca. 1% air saturation), cytochrome bd is induced QcrB subunit has been recently shown to be the target of
in M. smegmatis as the transition to anaerobiosis is novel imidazo[1,2-a]pyridine inhibitors that are active
approached (67), a value that is 10-fold lower than that against M. tuberculosis at low micromolar concen-
observed in E. coli (ca. 10% air saturation). In M. tu- trations (73).
berculosis, cytochrome bd is upregulated in the early As first reported by Niebisch and Bott (72), the QcrC
stages of NRP-1 (i.e., decreasing oxygen) (68), in NRP-2 subunit (cytochrome c1) harbors two heme-binding
(18), and in response to nitric oxide (NO) (18). Intrigu- signatures (CXXCH motifs) for c-type cytochromes
ingly, mRNA levels of cydA increased transiently about (e.g., CVSCH and CASCH), suggesting a covalent di-
7-fold after 30 days in mouse lungs (18). heme (bcc) configuration. Mycobacterial genomes do
Mycobacteria appear to adopt a strategy whereby not appear to harbor genes for either a soluble or
downregulation or slowing of metabolism occurs as membrane-bound cytochrome c, suggesting that the bcc
cells enter NRP-1 and NRP-2. Transcriptional analysis complex interacts directly with the aa3-type cytochrome
of M. tuberculosis in the macrophage (phagosomal c oxidase to achieve electron transfer and function as a
environment) has revealed that NDH-1, menaquinol- supercomplex. Niebisch and Bott (72, 74) suggest a
cytochrome c oxidoreductase, and the ATP synthase are supercomplex mechanism for C. glutamicum in which
all downregulated when compared to cells growing ex- the heme group of QcrC takes over the function of a
ponentially, suggesting that there is a reduced need for separate cytochrome c in electron transfer to cytochrome
energy generation during bacteriostasis, i.e., the growth aa3 oxidase, forging a close contact relationship between
state of intraphagosomal M. tuberculosis (19). Consistent cytochrome c1 and the CuA site in the subunit II of cy-
with these observations is the repression of these operons tochrome aa3 oxidase. Supercomplexes between bc1
during starvation. In contrast, FRD, nitrate reductase, and and aa3 have been reported for C. glutamicum (72),
NDH-2 are all upregulated under these conditions (19). Paracoccus denitrificans (75), and thermophilic strain
While these proteins do not appear to contribute to in- PS3 (76). A functional association between bcc and aa3
creased energy conservation, it has been suggested that has been shown for the complex in the M. smegmatis
they may play a pivotal role in the recycling of NAD+ as a family (71).
result of β-oxidation of fatty acids (19). The aa3-type cytochrome c oxidase in M. tubercu-
losis appears to be encoded by four genes, ctaBCDE
(Table 1). The ctaE gene is located immediately up-
Complex III and IV of the M. tuberculosis stream of the qcrCAB operon, but the other three genes
Electron Transport Chain are located in three different locations on the chromo-
M. tuberculosis harbors genes for a cytochrome c some and in some cases appear to be in operons with
pathway consisting of a menaquinol-cytochrome c oxi- other genes (Table 1). The four subunits of the M.
doreductase termed the bc1 complex (encoded by the tuberculosis aa3-type cytochrome c oxidase include CtaB

12 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

(a cytochrome c oxidase assembly factor), the catalytic and cydDC form two discrete operons, cydDC mutants
subunits CtaD (cytochrome c oxidase subunit I con- are defective in cytochrome bd assembly, and the
taining heme a, a3, and CuB) and CtaC (cytochrome c periplasmic space is more oxidized in the mutant versus
oxidase, subunit II location of copper A [CuA]), and wild type (79). In E. coli, the CydDC protein (ABC
CtaE (subunit III). For every two electrons passing transporter) has been reported to pump glutathione and
through complex IV, four protons are taken up from the cysteine into the periplasm to maintain redox homeo-
cytoplasm and two are released into the periplasm. The stasis (80). The role of the cydDC genes in mycobacteria
oxidase acts as a proton pump with a stoichiometry of is unknown, but evidence exists that CydDC plays a role
2H+/2e− (Fig. 2). during mycobacterial persistence in vivo. A cydC mutant
The bc1-aa3 pathway appears to be the major respira- of M. tuberculosis showed reduced ability to survive the
tory route in mycobacteria under standard aerobic transition from acute to chronic infection in mice (18),
culturing conditions (70). Matsoso et al. (70) have dem- and Dhar and McKinney have reported that CydC plays
onstrated that disruption of this pathway in M. smegmatis a role in mycobacterial persistence in INH-treated mice
is accompanied by a constitutive upregulation of the cy- (81).
tochrome bd-type menaquinol oxidase. In M. tuberculo-
sis, the bc1-aa3 pathway is essential for growth, suggesting
an inability of this bacterium to adapt in a manner anal- ELECTRON ACCEPTOR UTILIZATION
ogous to M. smegmatis (70). The aa3 branch is proposed UNDER HYPOXIA
to contain three ctaD alleles in M. smegmatis versus the In the absence of oxygen, alternative electron acceptors
one in M. tuberculosis, suggesting the existence of alter- (e.g., nitrate and fumarate) are available for mycobac-
native isoforms of cytochrome c oxidase in M. smegmatis terial metabolism, but none of these electron acceptors
(67). has been shown to support growth of mycobacterial
species. These potential electron acceptors may play an
Cytochrome bd-Type Oxidase important role in the disposal of reducing equivalents in
M. tuberculosis and other mycobacterial species harbor the absence of oxygen.
genes for the cytochrome bd-type menaquinol oxidase NO is generated in large amounts within the macro-
(cydAB) (67) (Table 1). The cytochrome bd branch is phages and restricts the growth of M. tuberculosis. Ni-
bioenergetically less efficient (non-proton-translocating) trate can be produced by oxidation of NO and is an
and is synthesized at low oxygen tensions in myco- alternative source of nitrogen for bacteria within the
bacteria (67). In addition to cytochrome bd, the M. human host. Early work in E. coli had suggested that
smegmatis respiratory chain has been proposed to con- narK was involved only in nitrite export (82), so the
tain a third possible respiratory branch terminating in homologous narK2 in M. tuberculosis was annotated as
the YthAB (bd-type) menaquinol oxidase (67). The ex- a “nitrite extrusion protein.” More recent work with an
istence of two cytochrome bd-type oxidases (I and II) is E. coli narK narU double mutant indicated that the two
not unprecedented in bacteria (77), and recent work has proteins could transport nitrate into and nitrite out of
reported that cytochrome bd-II in E. coli is able to the cell (83, 84). In M. tuberculosis, four genes, narK1
generate a PMF with a H+/e− ratio of 1.0 (78). Cyto- through narK3 and narU, are homologous to narK and
chrome bd mutants of E. coli have been shown to have a narU (Table 1). Since M. tuberculosis is unable to reduce
pleiotropic phenotype: they are sensitive to H2O2, nitric nitrite, which could accumulate to toxic levels, it must be
oxide, temperature, and iron (III) chelators and are un- exported out of the cell. The M. tuberculosis narK2 was
able to exit from stationary phase and resume aerobic shown to complement this E. coli double mutant, sup-
growth at 37°C (77). A cydA mutant of M. smegmatis porting a role for narK2 in nitrate reduction by coding
has been generated, but the phenotypes are very subtle for a transporter of nitrate into and nitrite out of the cell
compared to E. coli. The M. smegmatis cydA mutant (85). Nitrate reduction by M. tuberculosis is regulated by
showed a reduced growth rate at 0.5 to 1% air satura- control of nitrate transport into the cell by NarK2. It is
tion and also a 10-fold difference in CFU after 140 h of proposed that NarK2 senses the redox state of the cell,
growth in the presence of cyanide at 21% air saturation possibly by monitoring the flow of electrons to cyto-
when cocultured with wild-type M. smegmatis (67). chrome oxidase, and adjusts its activity so that nitrate is
The cydAB genes appear to be in an operon with transported under reducing, but not under oxidizing,
cydDC in mycobacterial species (Table 1), an arrange- conditions (86). Inhibition of nitrate transport by oxy-
ment similar to that found in Bacillus. In E. coli, cydAB gen has been documented in other bacteria (87). It is

ASMscience.org/MicrobiolSpectrum 13
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

intriguing that M. tuberculosis, classified as an obligate of the cell such that nitrate is transported into the cell
aerobe, should have such intricate control of an anaer- under reducing, but not oxidizing, conditions. Lastly,
obic enzyme system. Transcription of narK2 is controlled Tan et al. (92) proposed a role for nitrate reductase in
by DosR/DevR, which responds to O2, NO, and CO (88– protecting M. tuberculosis from mild acid challenge under
90). Both the transcription of the narK2 gene and the hypoxic conditions.
activity of NarK2 are controlled by similar signals (86). M. tuberculosis is unable to reduce nitrite, and the
M. tuberculosis contains genes (narGHJI, Rv1161– genome was proposed to harbor genes for a putative
1164) that code for a putative membrane-bound mo- ferredoxin-dependent nitrite reductase (NirA) and a NAD
lybdenum-containing nitrate reductase complex similar (P)H nitrite reductase (NirBD). However, a subsequent
to the corresponding narGHJI operon of E. coli study has reported that the nirA gene is misannotated and
(Table 1). Moreover, the narGHJI operon of M. tu- is in fact sirA, a sulfite reductase shown to be essential for
berculosis is able to functionally complement a nar growth on sulfate or sulfite as the sole sulfur source (93)
mutant of E. coli to grow on glycerol and reduce nitrate (Table 1). The nirBD genes are operonic and nonessential
anaerobically (85). Importantly, however, the expres- for growth, and unlike the narGHJI genes, no role in
sion of the narGHJI operon in M. tuberculosis is not resistance to acid stress under hypoxia was noted for
upregulated in response to either hypoxia or stationary NirBD (92). These data support the proposal that nitrite
phase (85). Sohaskey and Wayne demonstrated that produced by nitrate reductase is not converted to am-
overexpression of recombinant M. tuberculosis nitrate monium, but instead is exported from the cell by NarK2.
reductase in either M. tuberculosis or M. smegmatis (low The evolution of H2 is a mechanism commonly em-
nitrate reductase activity) does not confer the ability of ployed by anaerobic bacteria to recycle reducing
these cells to grow anaerobically; i.e., no growth of ei- equivalents obtained from anaerobic degradation of or-
ther species is observed with nitrate anaerobically, even ganic substrates. However, even strictly aerobic bacteria
though the nitrate reductase activity of whole cells have been shown to produce H2 under anaerobic
increases (85). The genome of M. tuberculosis also lacks conditions (94). In the family of Mycobacteriaceae, two
orthologs of transcription regulator FNR which, in potential hydrogen-evolving hydrogenases are present.
combination with NarL, are responsible for the tran- One belongs to group 2 cytoplasmic bidirectional
scriptional activation of the narGHJI operon by anaer- hydrogenases and can be found in some slow-growing
obic conditions in E. coli (21). A putative NarL (e.g., M. marinum and M. kansasii) and fast-growing
(Rv0884c) has been identified in M. tuberculosis, but the (e.g., M. smegmatis) mycobacteria. In M. smegmatis, this
promoter of the narGHJI lacks consensus-like binding enzyme is termed Hyd3 and has been shown to respond
sites for this regulatory protein. Based on these obser- strictly to oxygen-limiting conditions with up to a 50-
vations, it is apparent that this enzyme does not support fold increase in gene expression (16). The second type is a
anaerobic growth of mycobacteria, and therefore the yet uncharacterized enzyme that shows homology to
role of this enzyme in the physiology of mycobacteria is membrane-bound group 4 hydrogenases. This putative
unclear. Given the proposed membrane-bound location formate-hydrogen lyase is enocoded in M. tuberculosis
of the enzyme and the proton-translocating activity via a (viz., Rv0082 to Rv0087) and is upregulated during in-
redox loop of the E. coli enzyme, perhaps the primary fection of human macrophage-like THP-1 cells (95).
role of the mycobacterial enzyme is to generate a PMF Moreover, the transcription of the early genes of the
when the concentration of oxygen is low, and hence its hycP/hycQ-containing operon was shown to be up-
activity increases but not its expression. regulated during anaerobic adaptation in several studies
An alternative role for nitrate reductase may be main- (44, 68, 90, 96, 97). This gene cluster shows homology to
taining the redox balance of the cell during conditions of components of hydrogenase 4 and 3 complexes of E. coli,
hypoxia. Sohaskey (91) has reported that exogenously the latter of which has been shown to catalyze hydrogen
supplied nitrate has no effect on long-term persistence evolution at acidic pH (98). Upon closer inspection, the
during gradual oxygen depletion but played an important predicted [NiFe]-hydrogenase of M. tuberculosis appears
role during rapid adaptation to hypoxia (<18 h). This to form part of an Ehr (Ech hydrogenase-related) com-
effect required a functional nitrate reductase, suggesting plex (99, 100). Ehr enzymes lack the cysteine residues
that nitrate reduction may play a role in protecting cells needed to ligate a [NiFe] center, which is the catalytic
during sudden changes in oxygen concentration, leading center of hydrogenases. Additionally, no maturation
to disruption of aerobic respiration. The same author (86) factors to synthesize these complex centers are present in
has proposed a role for NarK2 in sensing the redox state M. tuberculosis.

14 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

Recently, it was shown that the operon Rv0081- M. tuberculosis generate a PMF in the order of −100 mV,
Rv0087 is positively regulated by the two two-component and the ATP synthase inhibitor TMC207 is bactericidal
signal transduction systems DosRS-DosT and MprAB toward these cells, demonstrating that the ATP synthase
and negatively regulated by Rv0081, a member of the still continues to function at relatively low PMF (6).
ArsR/SmtB family of metal-dependent transcriptional The F1F0-ATP synthase in M. tuberculosis and
regulators (101). DosRS-DosT is a redox-sensing regula- M. smegmatis has been shown to be essential for optimal
tory system that is important during the adaptation to growth (29, 103). In other bacteria, the F1F0-ATP
hypoxic conditions. No function has yet been proposed synthase is dispensable for growth on fermentable car-
for Ehr enzymes, but their homology to hydrogenases and bon sources (104, 105), where increased glycolytic flux
to proton-pumping type I NADH dehydrogenases points can compensate for the loss of oxidative phosphoryla-
to a role in energy metabolism. tion. This strategy does not appear to be exploited by
M. smegmatis: the F1F0-ATP synthase is essential for
growth even on fermentable substrates, suggesting that
ATP SYNTHESIS BY THE F1F0 ATP SYNTHASE ATP production from substrate level phosphorylation
In M. tuberculosis and other mycobacterial species, ATP alone, despite increased glycolytic flux, may be insuffi-
is synthesized via substrate level phosphorylation and cient to sustain growth of these bacteria (103). This may
oxidative phosphorylation using the membrane-bound be due to an extraordinarily high value for the amount
F1F0-ATP synthases (encoded by the atpBEFHAGDC of ATP required to synthesize a mycobacterial cell, a
operon, Rv1304-1311) (Table 1). In M. tuberculosis, possibility that requires further investigation (106). Al-
the atp operon is downregulated during growth in ternatively, in conjunction with a high ATP demand for
macrophages (19), in the mouse lung, and in cells ex- growth, the ATP synthase may be an obligatory re-
posed to NO or hypoxia (18). The atp operon of M. bovis quirement for the oxidation of NADH by providing a
BCG and M. smegmatis is downregulated in response to sink for translocated protons during NADH oxidation
slow growth rate (16, 45). When slow-growing cells of coupled to oxygen reduction (103). Such strict cou-
M. smegmatis (70 h doubling time), with low levels of atp pling would imply that mycobacteria do not support
operon expression, are exposed to hypoxia (0.6% oxy- uncoupled respiration; either they lack a conduit for
gen saturation), the atp operon is upregulated 3-fold, proton reentry in the absence of the F1F0-ATP synthase
suggesting an important role for this enzyme during ad- or they are unable to adjust the proton permeability of
aptation to hypoxia (16). the cytoplasmic membrane to allow a futile cycle of
The F1F0-ATP synthase catalyzes ATP synthesis by protons to operate. In this context, the cytoplasmic
utilizing the electrochemical gradient of protons to gen- membrane of M. smegmatis has been shown to be ex-
erate ATP from ADP and inorganic phosphate (Pi) and tremely impermeable to protons (107). The ATP syn-
operates under conditions of a high PMF and low intra- thase of the close mycobacterial phylogenetic relative
cellular ATP. The enzyme is also capable of working as an C. glutamicum is nonessential for growth on ferment-
ATPase under conditions of high intracellular ATP and an able carbon sources, and Δatp mutants of this bacterium
overall low PMF (102). As an ATPase, the enzyme show enhanced rates of glucose uptake, oxygen con-
hydrolyzes ATP while pumping protons from the cyto- sumption, and excretion of pyruvate into the growth
plasm to the outside of the cell. The ATP synthase of medium, suggesting that substrate level phosphorylation
mycobacteria has been studied in detail at a biochemical alone can sustain growth of this bacterium (108).
level in M. phlei and shown to exhibit latent ATPase ac- Several new antitubercular compounds have been
tivity (10). ATPase activity could be activated by trypsin reported that target oxidative phosphorylation in my-
treatment and magnesium ions, but the mechanism of cobacteria (12, 109, 110). The most promising com-
activation was not elucidated. Recent experiments with pounds clinically, the diarylquinolines, have been shown
inverted membrane vesicles of M. bovis BCG and to target the F1F0-ATP synthase and inhibit ATP syn-
M. smegmatis demonstrate latent ATPase activity that thesis (109, 111, 112). Genome sequencing of both
could be activated by methanol and the PMF, suggesting M. tuberculosis and M. smegmatis mutants that are re-
regulation by the epsilon subunit and ADP inhibition (9). sistant to diarylquinolines (i.e., TMC207) revealed that
The reason for the extreme latency in ATP hydrolysis the target of these compounds is the oligomeric c ring
of the mycobacterial ATP synthase is unknown but (encoded by atpE) of the enzyme (109, 113, 114). The
may represent an adaptation to function at low PMF purified c ring from M. smegmatis binds TMC207 with a
and under hypoxia. Hypoxic nonreplicating cells of KD of 500 nm, and modeling/docking and kinetic studies

ASMscience.org/MicrobiolSpectrum 15
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

suggest that TMC207 blocks rotary movement of the c WhiB3 has been implicated in sensing of oxygen and
ring during catalysis by mimicking key residues in the redox state by mycobacteria. Singh and colleagues re-
proton transfer chain (115, 116). Further investigations port that WhiB3 contains a 4Fe-4S cluster, which binds
with inverted membrane vesicles of M. smegmatis and NO (121). Furthermore, in the presence of oxygen, the
TMC207 have revealed that TMC207 acts indepen- WhiB3 [4Fe-4S]2+ cluster is degraded first to a [3Fe-4S]+
dently of the PMF and that electrostatic forces play an cluster, then a [2Fe-2S]2+ cluster and subsequently lost
important role in the interaction of the drug with the altogether, in a mechanism reminiscent of the one found
ATP synthase (116). in the E. coli oxygen sensor FNR (121, 122). Apo-
When mycobacterial cells (growing or nongrowing) WhiB3 was shown to have protein disulfide reductase
are treated with TMC207, time-dependent (not dose- activity, and it has been proposed that loss of the Fe-S
dependent) killing is observed (109). The mechanism of cluster is required to gain this activity (123). WhiB3-
killing is not clear, but it does not involve the dissipation mediated response to the presence of oxygen therefore
of the membrane potential, which is lethal to all living may occur through direct control of the activity of
cells. A dose-dependent decrease in intracellular ATP has metabolic proteins or through modification of tran-
been observed when M. tuberculosis cells are treated scriptional regulators (123). A role of WhiB3 in regu-
with TMC207 (111, 112), but these data do not explain lation of the transcriptional machinery in mycobacteria
cell death because mycobacterial cells can be depleted may be supported by the finding that WhiB3 interacts
of ATP and yet remain viable (117). TMC207 is bac- with the major sigma factor, SigA (RpoV) (124), but the
tericidal toward most species of mycobacteria (109) effect of this interaction on SigA activity is not known,
but is only bacteriostatic against M. avium (118) and and further study is required to understand the precise
M. smegmatis (M. Berney and G. M. Cook, unpublished role of WhiB3 in mediating any adaptation of myco-
data). Even when M. smegmatis was grown at a dou- bacteria to changes in oxygen tension.
bling time of 70 h in glycerol-limited continuous culture, A striking observation is that neither Dos nor WhiB3
TMC207 was bacteriostatic (Berney and Cook, unpub- controls any component of the electron transport ma-
lished data). The identification of the mechanisms chinery in M. tuberculosis. While the Dos system and
underlying this sensitivity will be important in under- WhiB3 are the most studied regarding the perception of
standing how TMC207 exerts its inhibitory effects on oxygen tension by mycobacteria, they are likely not the
mycobacterial cells. only systems used by these bacteria. For example, DosR of
M. tuberculosis H37Rv was shown not to be strictly re-
quired for survival of hypoxia in vitro (125). The authors
CONTROL OF ELECTRON TRANSPORT further found that expression of Dos regulon genes in re-
CHAIN EXPRESSION AND sponse to hypoxia was transient, with expression of about
OXIDATIVE PHOSPHORYLATION half of the 50 DosR-dependent genes returning to baseline
Mycobacteria are obligate aerobes and as such have to levels within 24 h of hypoxia. In contrast, a set of 230 genes
possess mechanisms to detect the ambient oxygen ten- was significantly upregulated at 4 and 7 days of hypoxia,
sion to enable them to adapt to changes in oxygen but not initially, and they were termed the enduring hy-
availability by adjusting their metabolism accordingly. poxic response (EHR) (125). Induction of the EHR was
Furthermore, gradual depletion of oxygen has been independent of DosR, suggesting that other sensory and
implicated in entry of M. tuberculosis into nonrepli- regulatory mechanisms must exist to signal prolonged
cating persistence and latency (119). The mechanisms by exposure to hypoxia. Strikingly, the EHR genes contained
which mycobacteria sense oxygen have been extensively an unusually high number of regulatory genes (FurA,
studied, and the two major sensory systems known to FurB, PhoP, three WhiB family members, and two ECF
date are the DosT/DosS/DosR system and WhiB3. sigma factors, SigH and SigE), but it is not yet known
The dormancy survival, or Dos, system consists of the which of these, if any, are involved in entry into EHR
three proteins DosR (or DevR), DosS (or DevS), and (125). A study into the effect of addition of cyclic AMP
DosT. DosR has been shown to act as a transcriptional (cAMP) to growing cultures of M. bovis BCG or M. tu-
regulator, which is responsible for the induction of berculosis H37Rv found that the number of genes affected
nearly all hypoxia-induced genes of M. tuberculosis by cAMP was larger under low oxygen, CO2-enriched
(96). In addition to hypoxia, the Dos-regulon has also conditions than under ambient air (126). The authors
been shown to be activated by exposure of M. tubercu- proposed that cAMP may be used by mycobacteria as a
losis to NO or CO (88, 90, 120). signaling molecule in response to hypoxic conditions.

16 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

However, the signal leading to cAMP synthesis, and which Squares R, Squares S, Stevens K, Taylor K, Whitehead S, Woodward JR,
adenylyl cyclase might catalyze this reaction under hyp- Barrell BG. 2001. Massive gene decay in the leprosy bacillus. Nature 409:
1007–1011.
oxia, remains unknown. The regulator of cydABDC ex-
14. Miesel L, Weisbrod TR, Marcinkeviciene JA, Bittman R, Jacobs WR
pression in mycobacteria has not been identified. Several Jr. 1998. NADH dehydrogenase defects confer isoniazid resistance and
regulators have been implicated in controlling the expres- conditional lethality in Mycobacterium smegmatis. J Bacteriol 180:2459–
sion of cyd expression, i.e., SenX3-RegX3 (127) and the 2467.
cAMP receptor protein (CRP) (128), but the signal trans- 15. Vilcheze C, Weisbrod TR, Chen B, Kremer L, Hazbon MH, Wang F,
Alland D, Sacchettini JC, Jacobs WR Jr. 2005. Altered NADH/NAD+
duction pathway or signals sensed remain to be elucidated. ratio mediates coresistance to isoniazid and ethionamide in mycobacteria.
Antimicrob Agents Chemother 49:708–720.
ACKNOWLEDGMENTS 16. Berney M, Cook GM. 2010. Unique flexibility in energy metabolism
Research in the authors’ laboratory is funded by the Health Re- allows mycobacteria to combat starvation and hypoxia. PLoS One 5:
search Council, Lottery Health, Marsden Fund, Royal Society e8614.
New Zealand, the Maurice Wilkins Center, and National Institute 17. Velmurugan K, Chen B, Miller JL, Azogue S, Gurses S, Hsu T,
of Health grant AI26170. Glickman M, Jacobs WR Jr, Porcelli SA, Briken V. 2007. Mycobacterium
tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected
host cells. PLoS Pathog 3:e110.
REFERENCES 18. Shi L, Sohaskey CD, Kana BD, Dawes S, North RJ, Mizrahi V,
1. Segal W, Bloch H. 1956. Biochemical differentiation of Mycobacterium Gennaro ML. 2005. Changes in energy metabolism of Mycobacterium
tuberculosis grown in vivo and in vitro. J Bacteriol 72:132–141. tuberculosis in mouse lung and under in vitro conditions affecting aerobic
2. Brodie AF, Gutnik DL. 1972. Electron transport and oxidative phos- respiration. Proc Natl Acad Sci USA 102:15629–15634.
phorylation in microbial systems, p 599–681. In King TE, Klingenberg M 19. Schnappinger D, Ehrt S, Voskuil MI, Liu Y, Mangan JA, Monahan
(ed), Electron and Coupled Energy Transfer Systems, vol. 1B. Marcel IM, Dolganov G, Efron B, Butcher PD, Nathan C, Schoolnik GK.
Dekker, New York. 2003. Transcriptional adaptation of Mycobacterium tuberculosis within
3. Russell JB, Cook GM. 1995. Energetics of bacterial growth: balance of macrophages: insights into the phagosomal environment. J Exp Med 198:
anabolic and catabolic reactions. Microbiol Rev 59:48–62. 693–704.
4. Rao M, Streur TL, Aldwell FE, Cook GM. 2001. Intracellular pH 20. Betts JC, Lukey PT, Robb LC, McAdam RA, Duncan K. 2002.
regulation by Mycobacterium smegmatis and Mycobacterium bovis BCG. Evaluation of a nutrient starvation model of Mycobacterium tuberculosis
Microbiology 147:1017–1024. persistence by gene and protein expression profiling. Mol Microbiol 43:
5. Ishaque M. 1992. Energy generation mechanisms in the in vitro-grown 717–731.
Mycobacterium lepraemurium. Int J Lepr Other Mycobact Dis 60:61–70. 21. Unden G, Bongaerts J. 1997. Alternative respiratory pathways of
6. Rao SP, Alonso S, Rand L, Dick T, Pethe K. 2008. The protonmotive Escherichia coli: energetics and transcriptional regulation in response to
force is required for maintaining ATP homeostasis and viability of hyp- electron acceptors. Biochim Biophys Acta 1320:217–234.
oxic, nonreplicating Mycobacterium tuberculosis. Proc Natl Acad Sci 22. Iwata M, Lee Y, Yamashita T, Yagi T, Iwata S, Cameron AD, Maher
USA 105:11945–11950. MJ. 2012. The structure of the yeast NADH dehydrogenase (Ndi1) reveals
7. Berney M, Weimar MR, Heikal A, Cook GM. 2012. Regulation of overlapping binding sites for water- and lipid-soluble substrates. Proc Natl
proline metabolism in mycobacteria and its role in carbon metabolism Acad Sci USA 109:15247–15252.
under hypoxia. Mol Microbiol 84:664–681. 23. Feng Y, Li WF, Li J, Wang JW, Ge JP, Xu D, Liu YJ, Wu KQ, Zeng
8. Dimroth P, Cook GM. 2004. Bacterial Na+ - or H+ -coupled ATP QY, Wu JW, Tian CL, Zhou B, Yang MJ. 2012. Structural insight into the
synthases operating at low electrochemical potential. Adv Microb Physiol type-II mitochondrial NADH dehydrogenases. Nature 491:478–482.
49:175–218. 24. Melo AMP, Bandeiras TM, Teixeira M. 2004. New insights into type
9. Haagsma AC, Driessen NN, Hahn MM, Lill H, Bald D. 2010. ATP II NAD(P)H : quinone oxidoreductases. Microbiol Mol Biol Rev 68:603–
synthase in slow- and fast-growing mycobacteria is active in ATP synthesis 616.
and blocked in ATP hydrolysis direction. FEMS Microbiol Lett 313: 25. Lin SS, Gross U, Bohne W. 2011. Two internal type II NADH
68–74. dehydrogenases of Toxoplasma gondii are both required for optimal
10. Higashi T, Kalra VK, Lee SH, Bogin E, Brodie AF. 1975. Energy- tachyzoite growth. Mol Microbiol 82:209–221.
transducing membrane-bound coupling factor-ATPase from Mycobacte- 26. Griffin JE, Gawronski JD, Dejesus MA, Ioerger TR, Akerley BJ,
rium phlei. I. Purification, homogeneity, and properties. J Biol Chem 250: Sassetti CM. 2011. High-resolution phenotypic profiling defines genes
6541–6548. essential for mycobacterial growth and cholesterol catabolism. PLoS
11. Kerscher S, Drose S, Zickermann V, Brandt U. 2008. The three Pathog 7:e1002251.
families of respiratory NADH dehydrogenases. Results Probl Cell Differ 27. Yano T, Li LS, Weinstein E, Teh JS, Rubin H. 2006. Steady-state
45:185–222. kinetics and inhibitory action of antitubercular phenothiazines on My-
12. Weinstein EA, Yano T, Li LS, Avarbock D, Avarbock A, Helm D, cobacterium tuberculosis type-II NADH-menaquinone oxidoreductase
McColm AA, Duncan K, Lonsdale JT, Rubin H. 2005. Inhibitors of type II (NDH-2). J Biol Chem 281:11456–11463.
NADH:menaquinone oxidoreductase represent a class of antitubercular 28. McAdam RA, Quan S, Smith DA, Bardarov S, Betts JC, Cook FC,
drugs. Proc Natl Acad Sci USA 102:4548–4553. Hooker EU, Lewis AP, Woollard P, Everett MJ, Lukey PT, Bancroft GJ,
13. Cole ST, Eiglmeier K, Parkhill J, James KD, Thomson NR, Wheeler Jacobs WR Jr, Duncan K. 2002. Characterization of a Mycobacterium
PR, Honore N, Garnier T, Churcher C, Harris D, Mungall K, Basham D, tuberculosis H37Rv transposon library reveals insertions in 351 ORFs
Brown D, Chillingworth T, Connor R, Davies RM, Devlin K, Duthoy S, and mutants with altered virulence. Microbiology 148:2975–2986.
Feltwell T, Fraser A, Hamlin N, Holroyd S, Hornsby T, Jagels K, Lacroix 29. Sassetti CM, Boyd DH, Rubin EJ. 2003. Genes required for myco-
C, Maclean J, Moule S, Murphy L, Oliver K, Quail MA, Rajandream MA, bacterial growth defined by high density mutagenesis. Mol Microbiol 48:
Rutherford KM, Rutter S, Seeger K, Simon S, Simmonds M, Skelton J, 77–84.

ASMscience.org/MicrobiolSpectrum 17
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

30. Warman AJ, Rito TS, Fisher NE, Moss DM, Berry NG, O’Neill PM, 48. Weigoldt M, Meens J, Bange FC, Pich A, Gerlach GF, Goethe R. 2013.
Ward SA, Biagini GA. 2013. Antitubercular pharmacodynamics of Metabolic adaptation of Mycobacterium avium subsp. paratuberculosis
phenothiazines. J Antimicrob Chemother 68:869–880. to the gut environment. Microbiology 159:380–391.
31. Teh JS, Yano T, Rubin H. 2007. Type II NADH: menaquinone oxi- 49. Smith DA, Parish T, Stoker NG, Bancroft GJ. 2001. Characterization
doreductase of Mycobacterium tuberculosis. Infectious Disorders Drug of auxotrophic mutants of Mycobacterium tuberculosis and their potential
Targets 7:169–181. as vaccine candidates. Infect Immun 69:1142–1150.
32. Biagini GA, Viriyavejakul P, O’Neill PM, Bray PG, Ward SA. 2006. 50. Vignais PM, Colbeau A. 2004. Molecular biology of microbial
Functional characterization and target validation of alternative complex I hydrogenases. Curr Issues Mol Biol 6:159–188.
of Plasmodium falciparum mitochondria. Antimicrob Agents Chemother 51. Tamagnini P, Leitao E, Oliveira P, Ferreira D, Pinto F, Harris DJ,
50:1841–1851. Heidorn T, Lindblad P. 2007. Cyanobacterial hydrogenases: diversity,
33. Shirude PS, Paul P, Choudhury NR, Kedari C, Bandodkar B, Ugarkar regulation and applications. FEMS Microbiol Rev 31:692–720.
BG. 2012. Quinolinyl pyrimidines: potent inhibitors of NDH-2 as a novel 52. Constant P, Chowdhury SP, Hesse L, Pratscher J, Conrad R. 2011.
class of anti-TB agents. ACS Med Chem Lett 3:736–740. Genome data mining and soil survey for the novel group 5 [NiFe]-
34. Ordway D, Viveiros M, Leandro C, Bettencourt R, Almeida J, Martins hydrogenase to explore the diversity and ecological importance of pre-
M, Kristiansen JE, Molnar J, Amaral L. 2003. Clinical concentrations of sumptive high-affinity H(2)-oxidizing bacteria. Appl Environ Microbiol
thioridazine kill intracellular multidrug-resistant Mycobacterium tuber- 77:6027–6035.
culosis. Antimicrob Agents Chemother 47:917–922. 53. Constant P, Chowdhury SP, Pratscher J, Conrad R. 2010. Strep-
35. Amaral L, Kristiansen JE, Abebe LS, Millett W. 1996. Inhibition of tomycetes contributing to atmospheric molecular hydrogen soil uptake are
the respiration of multi-drug resistant clinical isolates of Mycobacterium widespread and encode a putative high-affinity [NiFe]-hydrogenase. En-
tuberculosis by thioridazine: potential use for initial therapy of freshly viron Microbiol 12:821–829.
diagnosed tuberculosis. J Antimicrob Chemother 38:1049–1053. 54. King GM. 2003. Uptake of carbon monoxide and hydrogen at envi-
36. Mogi T, Matsushita K, Murase Y, Kawahara K, Miyoshi H, Ui H, ronmentally relevant concentrations by mycobacteria. Appl Environ
Shiomi K, Omura S, Kita K. 2009. Identification of new inhibitors for Microbiol 69:7266–7272.
alternative NADH dehydrogenase (NDH-II). FEMS Microbiol Lett 291: 55. Kim YM, Hegeman GD. 1983. Oxidation of carbon monoxide by
157–161. bacteria. Int Rev Cytol 81:1–32.
37. Tian J, Bryk R, Itoh M, Suematsu M, Nathan C. 2005. Variant 56. Park SW, Hwang EH, Park H, Kim JA, Heo J, Lee KH, Song T, Kim E,
tricarboxylic acid cycle in Mycobacterium tuberculosis: identification of Ro YT, Kim SW, Kim YM. 2003. Growth of mycobacteria on carbon
alpha-ketoglutarate decarboxylase. Proc Natl Acad Sci USA 102:10670– monoxide and methanol. J Bacteriol 185:142–147.
10675. 57. Schryvers A, Lohmeier E, Weiner JH. 1978. Chemical and functional
38. Youmans AS, Millman I, Youmans GP. 1956. The oxidation of properties of the native and reconstituted forms of the membrane-bound,
compounds related to the tricarboxylic acid cycle by whole cells and aerobic glycerol-3-phosphate dehydrogenase of Escherichia coli. J Biol
enzyme preparations of Mycobacterium tuberculosis var. hominis. Chem 253:783–788.
J Bacteriol 71:565–570. 58. Boos W. 1998. Binding protein-dependent ABC transport system for
39. Cecchini G, Schroder I, Gunsalus RP, Maklashina E. 2002. Succinate glycerol 3-phosphate of Escherichia coli. Methods Enzymol 292:40–51.
dehydrogenase and fumarate reductase from Escherichia coli. Biochimica 59. Molenaar D, van der Rest ME, Drysch A, Yucel R. 2000. Functions of
Biophysica Acta 1553:140–157. the membrane-associated and cytoplasmic malate dehydrogenases in the
40. Baek SH, Li AH, Sassetti CM. 2011. Metabolic regulation of myco- citric acid cycle of Corynebacterium glutamicum. J Bacteriol 182:6884–
bacterial growth and antibiotic sensitivity. PLoS Biol 9:e1001065. 6891.
41. Unden G, Schirawski J. 1997. The oxygen-responsive transcriptional 60. Molenaar D, van der Rest ME, Petrovic S. 1998. Biochemical and
regulator FNR of Escherichia coli: the search for signals and reactions. genetic characterization of the membrane-associated malate dehydroge-
Mol Microbiol 25:205–210. nase (acceptor) from Corynebacterium glutamicum. Eur J Biochem 254:
42. Watanabe S, Zimmermann M, Goodwin MB, Sauer U, Barry CE 3rd, 395–403.
Boshoff HI. 2011. Fumarate reductase activity maintains an energized 61. Prasada Reddy TL, Suryanarayana Murthy P, Venkitasubramanian
membrane in anaerobic Mycobacterium tuberculosis. PLoS Pathog 7: TA. 1975. Respiratory chains of Mycobacterium smegmatis. Indian J
e1002287. Biochem Biophys 12:255–259.
43. Eoh H, Rhee KY. 2013. Multifunctional essentiality of succinate 62. Prasada Reddy TL, Suryanarayana Murthy P, Venkitasubramanian
metabolism in adaptation to hypoxia in Mycobacterium tuberculosis. TA. 1975. Variations in the pathways of malate oxidation and phos-
Proc Natl Acad Sci USA 110:6554–6559. phorylation in different species of mycobacteria. Biochim Biophys Acta
44. Bacon J, James BW, Wernisch L, Williams A, Morley KA, Hatch GJ, 376:210–218.
Mangan JA, Hinds J, Stoker NG, Butcher PD, Marsh PD. 2004. The 63. D’Mello R, Hill S, Poole RK. 1995. The oxygen affinity of cytochrome
influence of reduced oxygen availability on pathogenicity and gene bo’ in Escherichia coli determined by the deoxygenation of oxyleghemo-
expression in Mycobacterium tuberculosis. Tuberculosis (Edinb) 84: globin and oxymyoglobin: Km values for oxygen are in the submicro-
205–217. molar range. J Bacteriol 177:867–870.
45. Beste DJ, Laing E, Bonde B, Avignone-Rossa C, Bushell ME, 64. D’Mello R, Hill S, Poole RK. 1996. The cytochrome bd quinol oxidase
McFadden JJ. 2007. Transcriptomic analysis identifies growth rate mod- in Escherichia coli has an extremely high oxygen affinity and two oxygen-
ulation as a component of the adaptation of mycobacteria to survival binding haems: implications for regulation of activity in vivo by oxygen
inside the macrophage. J Bacteriol 189:3969–3976. inhibition. Microbiology 142:755–763.
46. Tanner JJ. 2008. Structural biology of proline catabolism. Amino 65. Cotter PA, Melville SB, Albrecht JA, Gunsalus RP. 1997. Aerobic
Acids 35:719–730. regulation of cytochrome d oxidase (cydAB) operon expression in
47. Menzel R, Roth J. 1981. Purification of the putA gene product. Escherichia coli: roles of Fnr and ArcA in repression and activation. Mol
A bifunctional membrane-bound protein from Salmonella typhimurium Microbiol 25:605–615.
responsible for the two-step oxidation of proline to glutamate. J Biol 66. Tseng CP, Hansen AK, Cotter P, Gunsalus RP. 1994. Effect of cell
Chem 256:9755–9761. growth rate on expression of the anaerobic respiratory pathway operons

18 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Energetics of Respiration and Oxidative Phosphorylation in Mycobacteria

frdABCD, dmsABC, and narGHJI of Escherichia coli. J Bacteriol 176: 85. Sohaskey CD, Wayne LG. 2003. Role of narK2X and narGHJI in
6599–6605. hypoxic upregulation of nitrate reduction by Mycobacterium tuberculosis.
67. Kana BD, Weinstein EA, Avarbock D, Dawes SS, Rubin H, Mizrahi J Bacteriol 185:7247–7256.
V. 2001. Characterization of the cydAB-encoded cytochrome bd oxidase 86. Sohaskey CD. 2005. Regulation of nitrate reductase activity in
from Mycobacterium smegmatis. J Bacteriol 183:7076–7086. Mycobacterium tuberculosis by oxygen and nitric oxide. Microbiology
68. Voskuil MI, Visconti KC, Schoolnik GK. 2004. Mycobacterium 151:3803–3810.
tuberculosis gene expression during adaptation to stationary phase and 87. Moir JW, Wood NJ. 2001. Nitrate and nitrite transport in bacteria.
low-oxygen dormancy. Tuberculosis (Edinb) 84:218–227. Cell Mol Life Sci 58:215–224.
69. Boshoff HI, Barry CE 3rd. 2005. Tuberculosis: metabolism and res- 88. Kumar A, Deshane JS, Crossman DK, Bolisetty S, Yan BS, Kramnik I,
piration in the absence of growth. Nat Rev Microbiol 3:70–80. Agarwal A, Steyn AJ. 2008. Heme oxygenase-1-derived carbon monoxide
induces the Mycobacterium tuberculosis dormancy regulon. J Biol Chem
70. Matsoso LG, Kana BD, Crellin PK, Lea-Smith DJ, Pelosi A, Powell D,
Dawes SS, Rubin H, Coppel RL, Mizrahi V. 2005. Function of the cyto- 283:18032–18039.
chrome bc1-aa3 branch of the respiratory network in mycobacteria and 89. Ohno H, Zhu G, Mohan VP, Chu D, Kohno S, Jacobs WR Jr, Chan J.
network adaptation occurring in response to its disruption. J Bacteriol 2003. The effects of reactive nitrogen intermediates on gene expression in
187:6300–6308. Mycobacterium tuberculosis. Cell Microbiol 5:637–648.
71. Megehee JA, Hosler JP, Lundrigan MD. 2006. Evidence for a cyto- 90. Voskuil MI, Schnappinger D, Visconti KC, Harrell MI, Dolganov
chrome bcc-aa3 interaction in the respiratory chain of Mycobacterium GM, Sherman DR, Schoolnik GK. 2003. Inhibition of respiration by nitric
smegmatis. Microbiology 152:823–829. oxide induces a Mycobacterium tuberculosis dormancy program. J Exp
Med 198:705–713.
72. Niebisch A, Bott M. 2001. Molecular analysis of the cytochrome
bc1-aa3 branch of the Corynebacterium glutamicum respiratory chain 91. Sohaskey CD. 2008. Nitrate enhances the survival of Mycobac-
containing an unusual diheme cytochrome c1. Arch Microbiol 175:282– terium tuberculosis during inhibition of respiration. J Bacteriol 190:2981–
294. 2986.
73. Abrahams KA, Cox JA, Spivey VL, Loman NJ, Pallen MJ, 92. Tan MP, Sequeira P, Lin WW, Phong WY, Cliff P, Ng SH, Lee BH,
Constantinidou C, Fernandez R, Alemparte C, Remuinan MJ, Barros D, Camacho L, Schnappinger D, Ehrt S, Dick T, Pethe K, Alonso S. 2010.
Ballell L, Besra GS. 2012. Identification of novel imidazo[1,2-a]pyridine Nitrate respiration protects hypoxic Mycobacterium tuberculosis against
inhibitors targeting M. tuberculosis QcrB. PLoS One 7:e52951. acid- and reactive nitrogen species stresses. PLoS One 5:e13356.
74. Niebisch A, Bott M. 2003. Purification of a cytochrome bc-aa3 93. Pinto R, Harrison JS, Hsu T, Jacobs WR Jr, Leyh TS. 2007. Sulfite
supercomplex with quinol oxidase activity from Corynebacterium reduction in mycobacteria. J Bacteriol 189:6714–6722.
glutamicum. Identification of a fourth subunity of cytochrome aa3 oxidase 94. Kuhn M, Steinbuchel A, Schlegel HG. 1984. Hydrogen evolution by
and mutational analysis of diheme cytochrome c1. J Biol Chem 278:4339– strictly aerobic hydrogen bacteria under anaerobic conditions. J Bacteriol
4346. 159:633–639.
75. Berry EA, Trumpower BL. 1985. Isolation of ubiquinol oxidase from 95. Fontan P, Aris V, Ghanny S, Soteropoulos P, Smith I. 2008. Global
Paracoccus denitrificans and resolution into cytochrome bc1 and cyto- transcriptional profile of Mycobacterium tuberculosis during THP-1 hu-
chrome c-aa3 complexes. J Biol Chem 260:2458–2467. man macrophage infection. Infect Immun 76:717–725.
76. Sone N, Sekimachi M, Kutoh E. 1987. Identification and properties of 96. Park HD, Guinn KM, Harrell MI, Liao R, Voskuil MI, Tompa M,
a quinol oxidase super-complex composed of a bc1 complex and cyto- Schoolnik GK, Sherman DR. 2003. Rv3133c/dosR is a transcription
chrome oxidase in the thermophilic bacterium PS3. J Biol Chem 262: factor that mediates the hypoxic response of Mycobacterium tuberculosis.
15386–15391. Mol Microbiol 48:833–843.
77. Poole RK, Cook GM. 2000. Redundancy of aerobic respiratory chains 97. Sherman DR, Voskuil M, Schnappinger D, Liao R, Harrell MI,
in bacteria? Routes, reasons and regulation. Adv Microb Physiol 43:165– Schoolnik GK. 2001. Regulation of the Mycobacterium tuberculosis
224. hypoxic response gene encoding alpha-crystallin. Proc Natl Acad Sci USA
78. Borisov VB, Murali R, Verkhovskaya ML, Bloch DA, Han H, Gennis 98:7534–7539.
RB, Verkhovsky MI. 2011. Aerobic respiratory chain of Escherichia coli is 98. Mnatsakanyan N, Bagramyan K, Trchounian A. 2004. Hydro-
not allowed to work in fully uncoupled mode. Proc Natl Acad Sci USA genase 3 but not hydrogenase 4 is major in hydrogen gas production by
108:17320–17324. Escherichia coli formate hydrogenlyase at acidic pH and in the presence of
79. Goldman BS, Gabbert KK, Kranz RG. 1996. The temperature- external formate. Cell Biochem Biophys 41:357–366.
sensitive growth and survival phenotypes of Escherichia coli cydDC and 99. Coppi MV. 2005. The hydrogenases of Geobacter sulfurreducens: a
cydAB strains are due to deficiencies in cytochrome bd and are corrected comparative genomic perspective. Microbiology 151:1239–1254.
by exogenous catalase and reducing agents. J Bacteriol 178:6348–6351. 100. Marreiros BC, Batista AP, Duarte AM, Pereira MM. 2013. A
80. Pittman MS, Robinson HC, Poole RK. 2005. A bacterial glutathione missing link between complex I and group 4 membrane-bound [NiFe]
transporter (Escherichia coli CydDC) exports reductant to the periplasm. hydrogenases. Biochimica Biophysica Acta 1827:198–209.
J Biol Chem 280:32254–32261. 101. He H, Bretl DJ, Penoske RM, Anderson DM, Zahrt TC. 2011.
81. Dhar N, McKinney JD. 2010. Mycobacterium tuberculosis persistence Components of the Rv0081-Rv0088 locus, which encodes a predicted
mutants identified by screening in isoniazid-treated mice. Proc Natl Acad formate hydrogenlyase complex, are coregulated by Rv0081, MprA, and
Sci USA 107:12275–12280. DosR in Mycobacterium tuberculosis. J Bacteriol 193:5105–5118.
82. Rowe JJ, Ubbink-Kok T, Molenaar D, Konings WN, Driessen AJ. 102. von Ballmoos C, Cook GM, Dimroth P. 2008. Unique rotary ATP
1994. NarK is a nitrite-extrusion system involved in anaerobic nitrate synthase and its biological diversity. Annu Rev Biophys 37:43–64.
respiration by Escherichia coli. Mol Microbiol 12:579–586. 103. Tran SL, Cook GM. 2005. The F1F0-ATP synthase of Mycobacte-
83. Clegg S, Yu F, Griffiths L, Cole JA. 2002. The roles of the polytopic rium smegmatis is essential for growth. J Bacteriol 187:5023–5028.
membrane proteins NarK, NarU and NirC in Escherichia coli K-12: two 104. Friedl P, Hoppe J, Gunsalus RP, Michelsen O, von Meyenburg
nitrate and three nitrite transporters. Mol Microbiol 44:143–155. K, Schairer HU. 1983. Membrane integration and function of the three
84. Jia W, Cole JA. 2005. Nitrate and nitrite transport in Escherichia coli. F0 subunits of the ATP synthase of Escherichia coli K12. EMBO J 2:99–
Biochem Soc Trans 33:159–161. 103.

ASMscience.org/MicrobiolSpectrum 19
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45
Cook et al.

105. Santana M, Ionescu MS, Vertes A, Longin R, Kunst F, Danchin A, 117. Frampton R, Aggio RB, Villas-Boas SG, Arcus VL, Cook GM. 2012.
Glaser P. 1994. Bacillus subtilis F0F1 ATPase: DNA sequence of the atp Toxin-antitoxin systems of Mycobacterium smegmatis are essential for
operon and characterization of atp mutants. J Bacteriol 176:6802–6811. cell survival. J Biol Chem 287:5340–5356.
106. Cox RA, Cook GM. 2007. Growth regulation in the mycobacterial 118. Lounis N, Gevers T, Van den Berg J, Vranckx L, Andries K. 2009.
cell. Curr Mol Med 7:231–245. ATP synthase inhibition of Mycobacterium avium is not bactericidal.
107. Tran SL, Rao M, Simmers C, Gebhard S, Olsson K, Cook Antimicrob Agents Chemother 53:4927–4929.
GM. 2005. Mutants of Mycobacterium smegmatis unable to grow at 119. Wayne LG, Sohaskey CD. 2001. Nonreplicating persistence of my-
acidic pH in the presence of the protonophore carbonyl cyanide cobacterium tuberculosis. Annu Rev Microbiol 55:139–163.
m-chlorophenylhydrazone. Microbiology 151:665–672. 120. Shiloh MU, Manzanillo P, Cox JS. 2008. Mycobacterium tubercu-
108. Koch-Koerfges A, Kabus A, Ochrombel I, Marin K, Bott M. 2012. losis senses host-derived carbon monoxide during macrophage infection.
Physiology and global gene expression of a Corynebacterium glutamicum Cell Host Microbe 3:323–330.
DeltaF1F0-ATP synthase mutant devoid of oxidative phosphorylation. 121. Singh A, Guidry L, Narasimhulu KV, Mai D, Trombley J, Redding
Biochimica Biophysica Acta 1817:370–380. KE, Giles GI, Lancaster JR Jr, Steyn AJ. 2007. Mycobacterium tubercu-
109. Andries K, Verhasselt P, Guillemont J, Gohlmann HW, Neefs JM, losis WhiB3 responds to O2 and nitric oxide via its [4Fe-4S] cluster and is
Winkler H, Van Gestel J, Timmerman P, Zhu M, Lee E, Williams P, de essential for nutrient starvation survival. Proc Natl Acad Sci USA 104:
Chaffoy D, Huitric E, Hoffner S, Cambau E, Truffot-Pernot C, Lounis N, 11562–11567.
Jarlier V. 2005. A diarylquinoline drug active on the ATP synthase of 122. Crack J, Green J, Thomson AJ. 2004. Mechanism of oxygen sensing
Mycobacterium tuberculosis. Science 307:223–227. by the bacterial transcription factor fumarate-nitrate reduction (FNR).
110. Dhiman RK, Mahapatra S, Slayden RA, Boyne ME, Lenaerts A, J Biol Chem 279:9278–9286.
Hinshaw JC, Angala SK, Chatterjee D, Biswas K, Narayanasamy P, 123. Suhail Alam M, Agrawal P. 2008. Matrix-assisted refolding
Kurosu M, Crick DC. 2009. Menaquinone synthesis is critical for and redox properties of WhiB3/Rv3416 of Mycobacterium tuberculosis
maintaining mycobacterial viability during exponential growth and re- H37Rv. Protein Expr Purif 61:83–91.
covery from non-replicating persistence. Mol Microbiol 72:85–97. 124. Steyn AJ, Collins DM, Hondalus MK, Jacobs WR Jr, Kawakami RP,
111. Koul A, Dendouga N, Vergauwen K, Molenberghs B, Vranckx L, Bloom BR. 2002. Mycobacterium tuberculosis WhiB3 interacts with
Willebrords R, Ristic Z, Lill H, Dorange I, Guillemont J, Bald D, Andries RpoV to affect host survival but is dispensable for in vivo growth. Proc
K. 2007. Diarylquinolines target subunit c of mycobacterial ATP synthase. Natl Acad Sci USA 99:3147–3152.
Nat Chem Biol 3:323–324. 125. Rustad TR, Harrell MI, Liao R, Sherman DR. 2008. The en-
112. Koul A, Vranckx L, Dendouga N, Balemans W, Van den Wyngaert I, during hypoxic response of Mycobacterium tuberculosis. PLoS One 3:
Vergauwen K, Gohlmann HW, Willebrords R, Poncelet A, Guillemont J, e1502.
Bald D, Andries K. 2008. Diarylquinolines are bactericidal for dormant 126. Gazdik MA, McDonough KA. 2005. Identification of cyclic AMP-
mycobacteria as a result of disturbed ATP homeostasis. J Biol Chem 283: regulated genes in Mycobacterium tuberculosis complex bacteria under
25273–25280. low-oxygen conditions. J Bacteriol 187:2681–2692.
113. Huitric E, Verhasselt P, Andries K, Hoffner SE. 2007. In vitro 127. Roberts G, Vadrevu IS, Madiraju MV, Parish T. 2011. Control
antimycobacterial spectrum of a diarylquinoline ATP synthase inhibitor. of CydB and GltA1 expression by the SenX3 RegX3 two compo-
Antimicrob Agents Chemother 51:4202–4204. nent regulatory system of Mycobacterium tuberculosis. PLoS One 6:
114. Huitric E, Verhasselt P, Koul A, Andries K, Hoffner S, Andersson e21090.
DI. 2010. Rates and mechanisms of resistance development in Mycobac- 128. Rickman L, Scott C, Hunt DM, Hutchinson T, Menendez MC,
terium tuberculosis to a novel diarylquinoline ATP synthase inhibitor. Whalan R, Hinds J, Colston MJ, Green J, Buxton RS. 2005. A member of
Antimicrob Agents Chemother 54:1022–1028. the cAMP receptor protein family of transcription regulators in Myco-
115. de Jonge MR, Koymans LH, Guillemont JE, Koul A, Andries K. bacterium tuberculosis is required for virulence in mice and controls
2007. A computational model of the inhibition of Mycobacterium tu- transcription of the rpfA gene coding for a resuscitation promoting factor.
berculosis ATPase by a new drug candidate R207910. Proteins 67:971– Mol Microbiol 56:1274–1286.
980. 129. Zhang YJ, Ioerger TR, Huttenhower C, Long JE, Sassetti CM,
116. Haagsma AC, Podasca I, Koul A, Andries K, Guillemont J, Lill H, Sacchettini JC, Rubin EJ. 2012. Global assessment of genomic regions
Bald D. 2011. Probing the interaction of the diarylquinoline TMC207 required for growth in Mycobacterium tuberculosis. PLoS Pathog 8:
with its target mycobacterial ATP synthase. PLoS One 6:e23575. e1002946.

20 ASMscience.org/MicrobiolSpectrum
Downloaded from www.asmscience.org by
IP: 129.82.52.133
On: Sat, 17 Mar 2018 19:24:45

Você também pode gostar