Unique structural and mechanistic properties of mycobacterial F-ATP synthases: Implications for drug design

https://doi.org/10.1016/j.pbiomolbio.2019.11.006Get rights and content

Abstract

The causative agent of Tuberculosis (TB) Mycobacterium tuberculosis (Mtb) encounters unfavourable environmental conditions in the lungs, including nutrient limitation, low oxygen tensions and/or low/high pH values. These harsh conditions in the host triggers Mtb to enter a dormant state in which the pathogen does not replicate and uses host-derived fatty acids instead of carbohydrates as an energy source. Independent to the energy source, the bacterium’s energy currency ATP is generated by oxidative phosphorylation, in which the F1FO-ATP synthase uses the proton motive force generated by the electron transport chain. This catalyst is essential in Mtb and inhibition by the diarylquinoline class of drugs like Bedaquilline, TBAJ-587, TBAJ-876 or squaramides demonstrated that this engine is an attractive target in TB drug discovery. A special feature of the mycobacterial F-ATP synthase is its inability to establish a significant proton gradient during ATP hydrolysis, and its latent ATPase activity, to prevent energy waste and to control the membrane potential. Recently, unique epitopes of mycobacterial F1FO-ATP synthase subunits absent in their prokaryotic or mitochondrial counterparts have been identified to contribute to the regulation of the low ATPase activity. Most recent structural insights into individual subunits, the F1 domain or the entire mycobacterial enzyme added to the understanding of mechanisms, regulation and differences of the mycobacterial F1FO-ATP synthase compared to other bacterial and eukaryotic engines. These novel insights provide the basis for the design of new compounds targeting this engine and even novel regimens for multidrug resistant TB.

Introduction

During infection, Mtb inhabits a wide range of intracellular and extracellular environments, which the pathogen has to address by redirecting its metabolic activity commensurate with either replicative growth or nonreplicative persistence (Rao et al., 2008). A fundamental feature in this adaptation is the ability of mycobacteria to respire, regenerate reducing equivalents, and to generate adenosine triphosphate (ATP) via oxidative phosphorylation by the enzyme ensemble of the electron transport chain (ETC). Mycobacteria harbor dehydrogenases to fuel the ETC, and two terminal respiratory oxidases an aa3-type cytochrome c oxidase (cyt-bc1-aa3) and a bacterial specific cytochrome bd-type menaquinol oxidase (cyt-bd). These are present for dioxygen reduction coupled to the generation of a proton motive force (PMF, the sum of proton-gradient and the membrane potential), which is finally used for the condensation of ADP + Pi to form ATP by the engine F1FO-ATP synthase (F-ATP synthase) (Cook et al., 2014). Despite being obligate aerobes, mycobacteria survive under low oxygen tension (hypoxia), by exiting the cell cycle and entering a dormant state. Hypoxic non-replicating Mtb displays a reduced pool of ATP, making them exquisitely sensitive to any further ATP depletion, and susceptible to drugs targeting the maintenance of ATP homeostasis (Rao et al., 2008). This observation suggests that drugs that inhibit oxidative phosphorylation could shorten the time of therapy for drug-resistant tuberculosis which is supported by the clinical use of Sirturo® (bedaquiline, BDQ)), an anti-TB drug targeting the MtF-ATP synthase (Andries et al., 2005).

In contrast to other prokaryotes, the mycobacterial F-ATP synthase has been shown to be essential for growth and survival, which is important for the identification of novel antitubercular targets and agents (Sassetti et al., 2003). Another special feature of the mycobacterial F-ATP synthase is its inability to establish a significant proton gradient during ATP hydrolysis, and its low or latent ATPase activity in the fast- or slow-growing form (Haagsma et al., 2010; Hotra et al., 2016). Recent genetic, biochemical and structural approaches identified that at least three subunits (α, γ and ε) contribute to the important enzymatic differences of mycobacterial F-ATP synthases in suppression of proton pumping and PMF formation (Ragunathan et al., 2017; Hotra et al., 2016; Joon et al., 2018), which is essential because dissipating the PMF is lethal to mycobacteria (Cook et al., 2014), as well as in ATP formation, employing them as potential drug targets.

Unraveling the structural details of the mycobacterial F-ATP synthase is not only required for identifying the mycobacterial specific structural features of the enzyme, but also to exploit these specific features for the development of new antibacterial agents against the enzyme. Previous attempts have been made to understand the structure of the isolated subunits ε and c (Preiss et al., 2015; Joon et al., 2018) (Fig. 1). Most recent developments in producing a stable and enzymatically active recombinant M. smegmatis α3β3γε complex (referred to as MsF1-ATPase throughout the text (Zhang et al., 2019)) as well as an active endogenous M. smegmatis F1FO ATP synthase (MsF-ATP synthase) (Kamariah et al., 2019) provided structural insights into the subunit ensemble and arrangement of this engine (Fig. 1). Like other F-ATP synthases, the mycobacterial assembly consists of a water soluble F1 part that synthesizes ATP and a membrane embedded FO part that allows proton translocation from the periplasmic to the cytosolic side. The F1 part contains the nucleotide-binding subunits α3β3, forming the catalytic and hexameric headpiece, which sits on top of the rotating central stalk subunits γε (Fig. 1). The FO complex (subunits a:b:b’:c9) includes a rotary c-ring of nine c subunits (Preiss et al., 2015), where the loop docks to the bottom of the N-terminal domain of subunit ε and the globular domain of γ (Hotra et al., 2016; Joon et al., 2018), which both rotate and enable the coupling to the F1 portion to transfer torque, derived by H+-transport via subunits a and c, to the catalytic α3β3-headpiece. The peripheral stalk subunits b and b’, whose N-termini are membrane-embedded, extend and become attached via subunit δ to the top of the F1 headpiece (Kamariah et al., 2019). Together the peripheral- and central stalks establish two connections between the F1 and FO domains.

The discovery of BDQ, which is active against Mtb and its multi drug-resistant (MDR) strains, validated MtF-ATP synthase for drug development for tuberculosis (Andries et al. 2005, 2014). Most recent medicinal chemistry campaigns resulted in a new generation of 3,5-dialkoxypyridine (DARQ) analogues of BDQ (Tong et al., 2017; Choi et al., 2017; Sutherland et al., 2018, 2019; Blaser et al., 2019). In addition, the discovery of squaramides (SQA; Tantry et al., 2017), the compounds 5228485 and 5220632 (Kumar et al., 2018) as potent ATP synthesis inhibitors underline the attractiveness of the mycobacterial F-ATP synthase as a drug target. Therefore, the implementation of specific drug-mediated inhibition of the mycobacterial enzyme could increase efficacy, possibly decrease treatment time of a rational drug combination, and kill MDR- and extensively drug-resistant (XDR) bacterial strains of Mtb. In this review, we focus on developments in the understanding of the structure, function and regulation of mycobacterial F-ATP synthases in the past few years, whose outcomes will provide a first step toward structure-based drug design for the development of novel MtF-ATP synthase inhibitors.

Section snippets

Mycobacterial subunit α reveals a C-terminal extension important in regulation

Nucleotide-binding and ATP synthesis occurs in the interface of a subunit α-β pair. The mycobacterial subunit α (549 amino acid) includes a unique C-terminal stretch of about 3.5 kDa (Mt514–549; according to Mtb numbering), which is not found in other bacteria, chloroplast or mitochondrial sequences (Ragunathan et al., 2017). Secondary structural prediction revealed that this additional stretch is either entirely unstructured like in M. smegmatis, M. phlei and M. ulcerans, while in M. bovis or

Implications of an inhibited state of mycobacterial subunit β

Although each α-β heterodimer comprises a catalytic site, the majority of residues that contribute to nucleotide binding reside on the β subunit. Each β subunit has a catalytic- and a ‘lever’ domain, including the C-terminal called DELSEED-region. During ATP synthesis and hydrolysis, the central stalk of an F-ATP synthase rotates clockwise and counterclockwise, respectively, relative to the surrounding α3β3-ring (Noji et al., 1997; Diez et al., 2004; Spetzler et al., 2009). Each of the three

Mycobacterial subunit γ represents a specific TB drug target

The 310-amino acid mycobacterial subunit γ consists of the N- and C-terminal α-helices, forming an antiparallel coiled coil domain that protrudes into the catalytic α3β3. The N-terminal amino acids 24–34 of the M. smegmatis γ subunit interact with the C-terminal domains of the α- and β-subunits, and act like a rigid body (Zhang et al., 2019). Like in the crystal structure of the poorly hydrolysing F1-ATPase of C. thermarum (Ferguson et al., 2016), this rigid-body region of subunit γ rotates

Variations of mycobacterial subunit ε, its interaction with the TB-drug BDQ and its traits for a novel drug target

Previous trypsin proteolysis of IMVs from M. smegmatis, proposed to cleave subunit ε (Haagsma et al., 2010), as well as of recombinant MsF1-ATPase, demonstrating rapid cleavage of ε (Zhang et al., 2019), stimulated ATP hydrolysis. The importance of the mycobacterial subunit ε (121 residues) and in particular its C-terminus in regulating ATP hydrolysis became defined by a deletion mutant of the last and conserved residue D121 (mutant ε1-120) which exuded increased ATP hydrolysis activity in IMVs

Proton-transfer via the a-c domain and mechanism of action of BDQ within subunit c

The two entirely membrane-embedded mycobacterial FO subunits a and c build the core for proton translocation from the periplasmic-to the cytoplasmic side. So far only the M. phlei c (Mpc) subunit structure has been determined at atomic resolution (Preiss et al., 2015; PDB ID: 4v1f). Nine c subunits form the c-ring (Fig. 1), leading to a proton-to-ATP ratio of 3.0, whereby each Mpc N-terminus forms an ɑ-helix at the inner ring and the C-terminal ɑ-helix forms the outer ring side. Both helices

The evolutionary variation within the elastic mycobacterial peripheral stalk subunits

The flexibility within the mycobacterial peripheral stalk subunits b, b’ and δ is proposed to smoothen transmission of power between the rotary c-ring and the F1 domain with significant conformational variability of the C-terminal part of subunit b, which is in proximity to subunit δ (Kamariah et al., 2019). The gene encoding the b-δ subunits of mycobacterial F-ATP synthase is unique, in that the putative b subunit (atpF) and δ subunit (atpH) genes were fused to form a single gene (atpH) (

Acknowledgements:

This work as well as the research scholarship of Chui-Fann Wong was supported by the National Research Foundation (NRF) Singapore, NRF Competitive Research Programme (CRP), Grant Award Number NRF–CRP18–2017–01; Lead-PI G.G.). We are grateful to Dr. S. S. M. Malathy (School of Biological Sciences, NTU) for the art work of Fig. 1, Fig. 4C as well as Dr. Nebojša Bogdanović (School of Biological Sciences, NTU) of Figure 5A.

References (55)

  • H.S. Sutherland et al.

    3, 5-Dialkoxypyridine analogues of bedaquiline are potent antituberculosis agents with minimal inhibition of the hERG channel

    Bioorg. Med. Chem.

    (2019)
  • M. Yoshida et al.

    Identification of an essential glutamic acid residue in the beta subunit of the adenosine triphosphatase from the thermophilic bacterium PS3

    J. Biol. Chem.

    (1981)
  • J. Zheng et al.

    Rapid inhibition of rat brain mitochondrial proton F0F1-ATPase activity by estrogens: comparison with Na+, K+-ATPase of procine cortex

    Eur. J. Pharmacol.

    (1999)
  • K. Andries

    A diarylquinoline drug active on the ATP synthase of Mycobacterium tuberculosis

    Science

    (2005)
  • K. Andries

    Acquired resistance of Mycobacterium tuberculosis to bedaquiline

    PLoS One

    (2014)
  • G. Biuković et al.

    Variations of subunit epsilon of the Mycobacterium tuberculosis F1FO ATP synthase and a novel model for mechanism of action of the tuberculosis drug TMC207

    Antimicrob. Agents Chemother.

    (2013)
  • A. Blaser et al.

    Structure-activity relationships for unit C pyridyl analogues of the tuberculosis drug bedaquiline

    Bioorg. Med. Chem.

    (2019)
  • N. Bogdanović et al.

    The structural features of Acetobacterium woodii F-ATP synthase reveal the importance of the unique subunit γ-loop in Na+ translocation and ATP synthesis

    FEBS J.

    (2019)
  • R.A. Capaldi et al.

    Structural changes in the gamma and epsilon subunits of the Escherichia coli F1FO-type ATPase during energy coupling

    J. Bioenerg. Biomembr.

    (1996)
  • G. Cingolani et al.

    Structure of the ATP synthase catalytic complex (F(1)) from Escherichia coli in an autoinhibited conformation

    Nat. Struct. Mol. Biol.

    (2011)
  • L. Colina-Tenorio et al.

    The peripheral stalk of rotary ATPase

    Front. Physiol.

    (2018)
  • G.M. Cook et al.

    Energetics of respiration and oxidative phosphorylation in mycobacteria

    Microbiol. Spectr.

    (2014)
  • M. Diez et al.

    Proton-powered subunit rotation in single membrane-bound F0F1-ATP synthase

    Nat. Struct. Mol. Biol.

    (2004)
  • T. Duncan

    Turbine enzyme’s structure in the crosshairs to target tuberculosis

    Proc. Natl. Acad. Sci. U.S.A.

    (2019)
  • S.A. Ferguson et al.

    Regulation of the thermoalkaliphilic F1-ATPase from Caldalkalibacillus thermarum

    Proc. Natl. Acad. Sci. U.S.A.

    (2016)
  • R.H. Fillingame

    Identification of the dicyclohexylcarbodiimide-reactive protein component of the adenosine 5’-triphosphate energy-transducing system of Escherichia coli

    J. Bacteriol.

    (1975)
  • H. Guo et al.

    Structure of a bacterial ATP synthase

    Elife

    (2019)
  • Cited by (0)

    View full text