151
|
Gahura O, Panicucci B, Váchová H, Walker JE, Zíková A. Inhibition of F 1 -ATPase from Trypanosoma brucei by its regulatory protein inhibitor TbIF 1. FEBS J 2018; 285:4413-4423. [PMID: 30288927 DOI: 10.1111/febs.14672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/19/2018] [Accepted: 10/01/2018] [Indexed: 12/30/2022]
Abstract
Hydrolysis of ATP by the mitochondrial F-ATPase is inhibited by a protein called IF1 . In the parasitic flagellate, Trypanosoma brucei, this protein, known as TbIF1 , is expressed exclusively in the procyclic stage, where the F-ATPase is synthesizing ATP. In the bloodstream stage, where TbIF1 is absent, the F-ATPase hydrolyzes ATP made by glycolysis and compensates for the absence of a proton pumping respiratory chain by translocating protons into the intermembrane space, thereby maintaining the essential mitochondrial membrane potential. We have defined regions and amino acid residues of TbIF1 that are required for its inhibitory activity by analyzing the binding of several modified recombinant inhibitors to F1 -ATPase isolated from the procyclic stage of T. brucei. Kinetic measurements revealed that the C-terminal portion of TbIF1 facilitates homodimerization, but it is not required for the inhibitory activity, similar to the bovine and yeast orthologs. However, in contrast to bovine IF1 , the inhibitory capacity of the C-terminally truncated TbIF1 diminishes with decreasing pH, similar to full length TbIF1 . This effect does not involve the dimerization of active dimers to form inactive tetramers. Over a wide pH range, the full length and C-terminally truncated TbIF1 form dimers and monomers, respectively. TbIF1 has no effect on bovine F1 -ATPase, and this difference in the mechanism of regulation of the F-ATPase between the host and the parasite could be exploited in the design of drugs to combat human and animal African trypanosomiases.
Collapse
Affiliation(s)
- Ondřej Gahura
- Institute of Parasitology, Biology Centre Czech Academy of Science, České Budějovice, Czech Republic
- The Medical Research Council Mitochondrial Biology Unit, University of Cambridge, UK
| | - Brian Panicucci
- Institute of Parasitology, Biology Centre Czech Academy of Science, České Budějovice, Czech Republic
| | - Hana Váchová
- Institute of Parasitology, Biology Centre Czech Academy of Science, České Budějovice, Czech Republic
| | - John E Walker
- The Medical Research Council Mitochondrial Biology Unit, University of Cambridge, UK
| | - Alena Zíková
- Institute of Parasitology, Biology Centre Czech Academy of Science, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| |
Collapse
|
152
|
Abstract
Tuberculosis (TB) is one of the oldest infections afflicting humans yet remains the number one infectious disease killer worldwide. Despite decades of experience treating this disease, TB regimens require months of multidrug therapy, even for latent infections. There have been important recent advances in treatment options across the spectrum of TB, from latent infection to extensively drug-resistant (XDR) TB disease. In addition, new, potent drugs are emerging out of the development pipeline and are being tested in novel regimens in multiple currently enrolling trials. Shorter, safer regimens for many forms of TB are now available or are in our near-term vision. We review recent advances in TB therapeutics and provide an overview of the upcoming clinical trials landscape that will help define the future of worldwide TB treatment.
Collapse
Affiliation(s)
- Jeffrey A Tornheim
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;
| | - Kelly E Dooley
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA; .,Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.,Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
153
|
Marimani M, Ahmad A, Duse A. The role of epigenetics, bacterial and host factors in progression of Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2018; 113:200-214. [PMID: 30514504 DOI: 10.1016/j.tube.2018.10.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/21/2018] [Accepted: 10/23/2018] [Indexed: 12/29/2022]
Abstract
Tuberculosis (TB) infection caused by Mycobacterium tuberculosis (Mtb) is still a persistent global health problem, particularly in developing countries. The World Health Organization (WHO) reported a mortality rate of about 1.8 million worldwide due to TB complications in 2015. The Bacillus Calmette-Guérin (BCG) vaccine was introduced in 1921 and is still widely used to prevent TB development. This vaccine offers up to 80% protection against various forms of TB; however its efficacy against lung infection varies among different geographical settings. Devastatingly, the development of various forms of drug-resistant TB strains has significantly impaired the discovery of effective and safe anti-bacterial agents. Consequently, this necessitated discovery of new drug targets and novel anti-TB therapeutics to counter infection caused by various Mtb strains. Importantly, various factors that contribute to TB development have been identified and include bacterial resuscitation factors, host factors, environmental factors and genetics. Furthermore, Mtb-induced epigenetic changes also play a crucial role in evading the host immune response and leads to bacterial persistence and dissemination. Recently, the application of GeneXpert MTB/RIF® to rapidly diagnose and identify drug-resistant strains and discovery of different molecular markers that distinguish between latent and active TB infection has motivated and energised TB research. Therefore, this review article will briefly discuss the current TB state, highlight various mechanisms employed by Mtb to evade the host immune response as well as to discuss some modern molecular techniques that may potentially target and inhibit Mtb replication.
Collapse
Affiliation(s)
- Musa Marimani
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Aijaz Ahmad
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, South Africa.
| | - Adriano Duse
- Clinical Microbiology and Infectious Diseases, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, South Africa
| |
Collapse
|
154
|
Bogdanović N, Sundararaman L, Kamariah N, Tyagi A, Bhushan S, Ragunathan P, Shin J, Dick T, Grüber G. Structure and function of Mycobacterium-specific components of F-ATP synthase subunits α and ε. J Struct Biol 2018; 204:420-434. [PMID: 30342092 DOI: 10.1016/j.jsb.2018.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/08/2018] [Accepted: 10/16/2018] [Indexed: 01/21/2023]
Abstract
The Mycobacterium tuberculosis (Mtb) F1FO-ATP synthase (α3:β3:γ:δ:ε:a:b:b':c9) is an essential enzyme that supplies energy for both the aerobic growing and the hypoxic dormant stage of the mycobacterial life cycle. Employing the heterologous F-ATP synthase model system αchi3:β3:γ we showed previously, that transfer of the C-terminal domain (CTD) of Mtb subunit α (Mtα514-549) to a standard F-ATP synthase α subunit suppresses ATPase activity. Here we determined the 3D reconstruction from electron micrographs of the αchi3:β3:γ complex reconstituted with the Mtb subunit ε (Mtε), which has been shown to crosstalk with the CTD of Mtα. Together with the first solution shape of Mtb subunit α (Mtα), derived from solution X-ray scattering, the structural data visualize the extended C-terminal stretch of the mycobacterial subunit α. In addition, Mtε mutants MtεR62L, MtεE87A, Mtε6-121, and Mtε1-120, reconstituted with αchi3:β3:γ provided insight into their role in coupling and in trapping inhibiting MgADP. NMR solution studies of MtεE87A gave insights into how this residue contributes to stability and crosstalk between the N-terminal domain (NTD) and the CTD of Mtε. Analyses of the N-terminal mutant Mtε6-121 highlight the differences of the NTD of mycobacterial subunit ε to the well described Geobacillus stearothermophilus or Escherichia coli counterparts. These data are discussed in context of a crosstalk between the very N-terminal amino acids of Mtε and the loop region of one c subunit of the c-ring turbine for coupling of proton-translocation and ATP synthesis activity.
Collapse
Affiliation(s)
- Nebojša Bogdanović
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Lavanya Sundararaman
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Neelagandan Kamariah
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Anu Tyagi
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Shashi Bhushan
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore; NTU Institute of Structural Biology, Nanyang Technological University, Republic of Singapore
| | - Priya Ragunathan
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Joon Shin
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Thomas Dick
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Republic of Singapore; Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, 225 Warren Street, Newark, NJ 07103, USA
| | - Gerhard Grüber
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore.
| |
Collapse
|
155
|
|
156
|
Abstract
Bedaquiline (BDQ), an inhibitor of the mycobacterial F1Fo-ATP synthase, has revolutionized the antitubercular drug discovery program by defining energy metabolism as a potent new target space. Several studies have recently suggested that BDQ ultimately causes mycobacterial cell death through a phenomenon known as uncoupling. The biochemical basis underlying this, in BDQ, is unresolved and may represent a new pathway to the development of effective therapeutics. In this communication, we demonstrate that BDQ can inhibit ATP synthesis in Escherichia coli by functioning as a H+/K+ ionophore, causing transmembrane pH and potassium gradients to be equilibrated. Despite the apparent lack of a BDQ-binding site, incorporating the E. coli Fo subunit into liposomes enhanced the ionophoric activity of BDQ. We discuss the possibility that localization of BDQ at F1Fo-ATP synthases enables BDQ to create an uncoupled microenvironment, by antiporting H+/K+ Ionophoric properties may be desirable in high-affinity antimicrobials targeting integral membrane proteins.
Collapse
|
157
|
A uniform cloning platform for mycobacterial genetics and protein production. Sci Rep 2018; 8:9539. [PMID: 29934571 PMCID: PMC6015033 DOI: 10.1038/s41598-018-27687-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/08/2018] [Indexed: 12/31/2022] Open
Abstract
Molecular research on mycobacteria relies on a multitude of tools for the genetic manipulation of these clinically important bacteria. However, a uniform set of vectors allowing for standardized cloning procedures is not available. Here, we developed a versatile series of mycobacterial vectors for gene deletion, complementation and protein production and purification. The vectors are compatible with fragment exchange (FX) cloning, a recently developed high-throughput cloning principle taking advantage of the type IIS restriction enzyme SapI and its capacity to generate sticky trinucleotide ends outside of its recognition sequence. FX cloning allows for the efficient cloning into an entry vector and the facile transfer of the sequenced insert into a variety of destination vectors. We generated a set of mycobacterial expression vectors spanning a wide range of expression strengths, tagging variants and selection markers to rapidly screen for the optimal expression construct in order to purify membrane proteins from the model organism Mycobacterium smegmatis. Further, we generated a series of suicide vectors containing two counterselection markers and used them to delete twenty genes encoding for potential drug efflux pumps in M. smegmatis. The vectors will further facilitate genetic and biochemical research on various mycobacterial species.
Collapse
|
158
|
Tomatidine Is a Lead Antibiotic Molecule That Targets Staphylococcus aureus ATP Synthase Subunit C. Antimicrob Agents Chemother 2018; 62:AAC.02197-17. [PMID: 29610201 DOI: 10.1128/aac.02197-17] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/27/2018] [Indexed: 12/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of deadly hospital-acquired infections. The discovery of anti-Staphylococcus antibiotics and new classes of drugs not susceptible to the mechanisms of resistance shared among bacteria is imperative. We recently showed that tomatidine (TO), a steroidal alkaloid from solanaceous plants, possesses potent antibacterial activity against S. aureus small-colony variants (SCVs), the notoriously persistent form of this bacterium that has been associated with recurrence of infections. Here, using genomic analysis of in vitro-generated TO-resistant S. aureus strains to identify mutations in genes involved in resistance, we identified the bacterial ATP synthase as the cellular target. Sequence alignments were performed to highlight the modified sequences, and the structural consequences of the mutations were evaluated in structural models. Overexpression of the atpE gene in S. aureus SCVs or introducing the mutation found in the atpE gene of one of the high-level TO-resistant S. aureus mutants into the Bacillus subtilis atpE gene provided resistance to TO and further validated the identity of the cellular target. FC04-100, a TO derivative which also possesses activity against non-SCV strains, prevents high-level resistance development in prototypic strains and limits the level of resistance observed in SCVs. An ATP synthesis assay allowed the observation of a correlation between antibiotic potency and ATP synthase inhibition. The selectivity index (inhibition of ATP production by mitochondria versus that of bacterial ATP synthase) is estimated to be >105-fold for FC04-100.
Collapse
|
159
|
Sutherland HS, Tong AST, Choi PJ, Conole D, Blaser A, Franzblau SG, Cooper CB, Upton AM, Lotlikar MU, Denny WA, Palmer BD. Structure-activity relationships for analogs of the tuberculosis drug bedaquiline with the naphthalene unit replaced by bicyclic heterocycles. Bioorg Med Chem 2018; 26:1797-1809. [PMID: 29482950 PMCID: PMC5933462 DOI: 10.1016/j.bmc.2018.02.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 12/22/2022]
Abstract
Replacing the naphthalene C-unit of the anti-tuberculosis drug bedaquiline with a range of bicyclic heterocycles of widely differing lipophilicity gave analogs with a 4.5-fold range in clogP values. The biological results for these compounds indicate on average a lower clogP limit of about 5.0 in this series for retention of potent inhibitory activity (MIC90s) against M.tb in culture. Some of the compounds also showed a significant reduction in inhibition of hERG channel potassium current compared with bedaquiline, but there was no common structural feature that distinguished these.
Collapse
Affiliation(s)
- Hamish S Sutherland
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Amy S T Tong
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Peter J Choi
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Daniel Conole
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Adrian Blaser
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612, USA
| | | | - Anna M Upton
- Global Alliance for TB Drug Development, 40 Wall St, New York, NY 10005, USA
| | - Manisha U Lotlikar
- Global Alliance for TB Drug Development, 40 Wall St, New York, NY 10005, USA
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Brian D Palmer
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
160
|
Krah A, Zarco-Zavala M, McMillan DGG. Insights into the regulatory function of the ɛ subunit from bacterial F-type ATP synthases: a comparison of structural, biochemical and biophysical data. Open Biol 2018; 8:170275. [PMID: 29769322 PMCID: PMC5990651 DOI: 10.1098/rsob.170275] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/24/2018] [Indexed: 01/07/2023] Open
Abstract
ATP synthases catalyse the formation of ATP, the most common chemical energy storage unit found in living cells. These enzymes are driven by an electrochemical ion gradient, which allows the catalytic evolution of ATP by a binding change mechanism. Most ATP synthases are capable of catalysing ATP hydrolysis to varying degrees, and to prevent wasteful ATP hydrolysis, bacteria and mitochondria have regulatory mechanisms such as ADP inhibition. Additionally, ɛ subunit inhibition has also been described in three bacterial systems, Escherichia coli, Bacillus PS3 and Caldalkalibacillus thermarum TA2.A1. Previous studies suggest that the ɛ subunit is capable of undergoing an ATP-dependent conformational change from the ATP hydrolytic inhibitory 'extended' conformation to the ATP-induced non-inhibitory 'hairpin' conformation. A recently published crystal structure of the F1 domain of the C. thermarum TA2.A1 F1Fo ATP synthase revealed a mutant ɛ subunit lacking the ability to bind ATP in a hairpin conformation. This is a surprising observation considering it is an organism that performs no ATP hydrolysis in vivo, and appears to challenge the current dogma on the regulatory role of the ɛ subunit. This has prompted a re-examination of present knowledge of the ɛ subunits role in different organisms. Here, we compare published biochemical, biophysical and structural data involving ɛ subunit-mediated ATP hydrolysis regulation in a variety of organisms, concluding that the ɛ subunit from the bacterial F-type ATP synthases is indeed capable of regulating ATP hydrolysis activity in a wide variety of bacteria, making it a potentially valuable drug target, but its exact role is still under debate.
Collapse
Affiliation(s)
- Alexander Krah
- School of Computational Sciences, Korea Institute for Advanced Study, 85 Hoegiro Dongdaemun-gu, Seoul 02455, Republic of Korea
| | - Mariel Zarco-Zavala
- Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Duncan G G McMillan
- Department of Biotechnology, Delft University of Technology, van der Maasweg 9, Delft 2629 HZ, The Netherlands
| |
Collapse
|
161
|
Zhou W, Faraldo-Gómez JD. Membrane plasticity facilitates recognition of the inhibitor oligomycin by the mitochondrial ATP synthase rotor. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:789-796. [PMID: 29630891 DOI: 10.1016/j.bbabio.2018.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/13/2018] [Accepted: 03/23/2018] [Indexed: 12/11/2022]
Abstract
Enzymes in the respiratory chain are increasingly seen as potential targets against multi-drug resistance of human pathogens and cancerous cells. However, a detailed understanding of the mechanism and specificity determinants of known inhibitors is still lacking. Oligomycin, for example, has been known to be an inhibitor of the membrane motor of the mitochondrial ATP synthase for over five decades, and yet little is known about its mode of action at the molecular level. In a recent breakthrough, a crystal structure of the S. cerevisiae c-subunit ring with bound oligomycin revealed the inhibitor docked on the outer face of the proton-binding sites, deep into the transmembrane region. However, the structure of the complex was obtained in an organic solvent rather than detergent or a lipid bilayer, and therefore it has been unclear whether this mode of recognition is physiologically relevant. Here, we use molecular dynamics simulations to address this question and gain insights into the mechanism of oligomycin inhibition. Our findings lead us to propose that oligomycin naturally partitions into the lipid/water interface, and that in this environment the inhibitor can indeed bind to any of the c-ring proton-carrying sites that are exposed to the membrane, thereby becoming an integral component of the proton-coordinating network. As the c-ring rotates within the membrane, driven either by downhill proton permeation or ATP hydrolysis, one of the protonated, oligomycin-bound sites eventually reaches the subunit-a interface and halts the rotary mechanism of the enzyme.
Collapse
Affiliation(s)
- Wenchang Zhou
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Room 5N307A, Bethesda, MD 20892, United States
| | - José D Faraldo-Gómez
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, 10 Center Drive, Room 5N307A, Bethesda, MD 20892, United States.
| |
Collapse
|
162
|
Iqbal IK, Bajeli S, Akela AK, Kumar A. Bioenergetics of Mycobacterium: An Emerging Landscape for Drug Discovery. Pathogens 2018; 7:E24. [PMID: 29473841 PMCID: PMC5874750 DOI: 10.3390/pathogens7010024] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) exhibits remarkable metabolic flexibility that enables it to survive a plethora of host environments during its life cycle. With the advent of bedaquiline for treatment of multidrug-resistant tuberculosis, oxidative phosphorylation has been validated as an important target and a vulnerable component of mycobacterial metabolism. Exploiting the dependence of Mtb on oxidative phosphorylation for energy production, several components of this pathway have been targeted for the development of new antimycobacterial agents. This includes targeting NADH dehydrogenase by phenothiazine derivatives, menaquinone biosynthesis by DG70 and other compounds, terminal oxidase by imidazopyridine amides and ATP synthase by diarylquinolines. Importantly, oxidative phosphorylation also plays a critical role in the survival of persisters. Thus, inhibitors of oxidative phosphorylation can synergize with frontline TB drugs to shorten the course of treatment. In this review, we discuss the oxidative phosphorylation pathway and development of its inhibitors in detail.
Collapse
Affiliation(s)
- Iram Khan Iqbal
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh 160036, India.
| | - Sapna Bajeli
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh 160036, India.
| | - Ajit Kumar Akela
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh 160036, India.
| | - Ashwani Kumar
- Council of Scientific and Industrial Research, Institute of Microbial Technology, Chandigarh 160036, India.
| |
Collapse
|
163
|
Joon S, Ragunathan P, Sundararaman L, Nartey W, Kundu S, Manimekalai MSS, Bogdanović N, Dick T, Grüber G. The NMR solution structure of Mycobacterium tuberculosis F-ATP synthase subunit ε provides new insight into energy coupling inside the rotary engine. FEBS J 2018; 285:1111-1128. [PMID: 29360236 DOI: 10.1111/febs.14392] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 11/30/2017] [Accepted: 01/18/2018] [Indexed: 11/27/2022]
Abstract
Mycobacterium tuberculosis (Mt) F1 F0 ATP synthase (α3 :β3 :γ:δ:ε:a:b:b':c9 ) is essential for the viability of growing and nongrowing persister cells of the pathogen. Here, we present the first NMR solution structure of Mtε, revealing an N-terminal β-barrel domain (NTD) and a C-terminal domain (CTD) composed of a helix-loop-helix with helix 1 and -2 being shorter compared to their counterparts in other bacteria. The C-terminal amino acids are oriented toward the NTD, forming a domain-domain interface between the NTD and CTD. The Mtε structure provides a novel mechanistic model of coupling c-ring- and ε rotation via a patch of hydrophobic residues in the NTD and residues of the CTD to the bottom of the catalytic α3 β3 -headpiece. To test our model, genome site-directed mutagenesis was employed to introduce amino acid changes in these two parts of the epsilon subunit. Inverted vesicle assays show that these mutations caused an increase in ATP hydrolysis activity and a reduction in ATP synthesis. The structural and enzymatic data are discussed in light of the transition mechanism of a compact and extended state of Mtε, which provides the inhibitory effects of this coupling subunit inside the rotary engine. Finally, the employment of these data with molecular docking shed light into the second binding site of the drug Bedaquiline. DATABASE Structural data are available in the PDB under the accession number 5YIO.
Collapse
Affiliation(s)
- Shin Joon
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| | - Priya Ragunathan
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| | - Lavanya Sundararaman
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| | - Wilson Nartey
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| | - Subhashri Kundu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Nebojša Bogdanović
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| | - Thomas Dick
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Gerhard Grüber
- Nanyang Technological University, School of Biological Sciences, Singapore, Singapore
| |
Collapse
|
164
|
Ismail NA, Omar SV, Joseph L, Govender N, Blows L, Ismail F, Koornhof H, Dreyer AW, Kaniga K, Ndjeka N. Defining Bedaquiline Susceptibility, Resistance, Cross-Resistance and Associated Genetic Determinants: A Retrospective Cohort Study. EBioMedicine 2018; 28:136-142. [PMID: 29337135 PMCID: PMC5835552 DOI: 10.1016/j.ebiom.2018.01.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/22/2017] [Accepted: 01/04/2018] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Bedaquiline (BDQ) is a novel agent approved for use in combination treatment of multi-drug resistant tuberculosis (MDR-TB). We sought to determine BDQ epidemiological cut-off values (ECVs), define and assess interpretive criteria against putative resistance associated variants (RAVs), microbiological outcomes and cross resistance with clofazimine (CFZ). METHODS A retrospective cohort study was conducted. Minimal inhibitory concentrations (MIC) to BDQ were determined using 7H9 broth microdilution (BMD) and MGIT960. RAVs were genetically characterised using whole genome sequencing. BDQ ECVs were determined using ECOFFinder and compared with 6-month culture conversion status and CFZ MICs. FINDINGS A total of 391 isolates were analysed. Susceptible and intermediate categories were determined to have MICs of ≤0.125μg/ml and 0.25μg/ml using BMD and ≤1μg/ml and 2μg/ml using MGIT960 respectively. Microbiological failures occurred among BDQ exposed patients with a non-susceptible BDQ MIC, an Rv0678 mutation and ≤2 active drug classes. The Rv0678 RAVs were not the dominant mechanism of CFZ resistance and cross resistance was limited to isolates with an Rv0678 mutation. INTERPRETATION Criteria for BDQ susceptibility are defined and will facilitate improved early detection of resistance. Cross- resistance between BDQ and CFZ is an emerging concern but in this study was primarily among those with an Rv0678 mutation.
Collapse
Affiliation(s)
- Nazir A Ismail
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa; Department of Medical Microbiology, University of Pretoria, Pretoria, South Africa.
| | - Shaheed V Omar
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Lavania Joseph
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Netricia Govender
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Linsay Blows
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Farzana Ismail
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa; Department of Medical Microbiology, University of Pretoria, Pretoria, South Africa
| | - Hendrik Koornhof
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Andries W Dreyer
- National Institute for Communicable Diseases, Centre for Tuberculosis, Johannesburg, South Africa
| | - Koné Kaniga
- Janssen Research & Development, Titusville, NJ, United States
| | - Norbert Ndjeka
- National Department of Health, Tuberculosis Control and Management Cluster, Pretoria, South Africa
| |
Collapse
|
165
|
Hards K, Cook GM. Targeting bacterial energetics to produce new antimicrobials. Drug Resist Updat 2018; 36:1-12. [DOI: 10.1016/j.drup.2017.11.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
|
166
|
Abstract
Mitochondria are the power stations of the eukaryotic cell, using the energy released by the oxidation of glucose and other sugars to produce ATP. Electrons are transferred from NADH, produced in the citric acid cycle in the mitochondrial matrix, to oxygen by a series of large protein complexes in the inner mitochondrial membrane, which create a transmembrane electrochemical gradient by pumping protons across the membrane. The flow of protons back into the matrix via a proton channel in the ATP synthase leads to conformational changes in the nucleotide binding pockets and the formation of ATP. The three proton pumping complexes of the electron transfer chain are NADH-ubiquinone oxidoreductase or complex I, ubiquinone-cytochrome c oxidoreductase or complex III, and cytochrome c oxidase or complex IV. Succinate dehydrogenase or complex II does not pump protons, but contributes reduced ubiquinone. The structures of complex II, III and IV were determined by x-ray crystallography several decades ago, but complex I and ATP synthase have only recently started to reveal their secrets by advances in x-ray crystallography and cryo-electron microscopy. The complexes I, III and IV occur to a certain extent as supercomplexes in the membrane, the so-called respirasomes. Several hypotheses exist about their function. Recent cryo-electron microscopy structures show the architecture of the respirasome with near-atomic detail. ATP synthase occurs as dimers in the inner mitochondrial membrane, which by their curvature are responsible for the folding of the membrane into cristae and thus for the huge increase in available surface that makes mitochondria the efficient energy plants of the eukaryotic cell.
Collapse
Affiliation(s)
- Joana S Sousa
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Edoardo D'Imprima
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Janet Vonck
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
167
|
Kang SJ, Todokoro Y, Bak S, Suzuki T, Yoshida M, Fujiwara T, Akutsu H. Direct assignment of 13C solid-state NMR signals of TF oF 1 ATP synthase subunit c-ring in lipid membranes and its implication for the ring structure. JOURNAL OF BIOMOLECULAR NMR 2018; 70:53-65. [PMID: 29197977 DOI: 10.1007/s10858-017-0158-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/28/2017] [Indexed: 06/07/2023]
Abstract
FoF1-ATP synthase catalyzes ATP hydrolysis/synthesis coupled with a transmembrane H+ translocation in membranes. The Fo c-subunit ring plays a major role in this reaction. We have developed an assignment strategy for solid-state 13C NMR (ssNMR) signals of the Fo c-subunit ring of thermophilic Bacillus PS3 (TFo c-ring, 72 residues), carrying one of the basic folds of membrane proteins. In a ssNMR spectrum of uniformly 13C-labeled sample, the signal overlap has been a major bottleneck because most amino acid residues are hydrophobic. To overcome signal overlapping, we developed a method designated as COmplementary Sequential assignment with MInimum Labeling Ensemble (COSMILE). According to this method, we generated three kinds of reverse-labeled samples to suppress signal overlapping. To assign the carbon signals sequentially, two-dimensional Cα(i+1)-C'Cα(i) correlation and dipolar assisted rotational resonance (DARR) experiments were performed under magic-angle sample spinning. On the basis of inter- and intra-residue 13C-13C chemical shift correlations, 97% of Cα, 97% of Cβ and 92% of C' signals were assigned directly from the spectra. Secondary structure analysis predicted a hairpin fold of two helices with a central loop. The effects of saturated and unsaturated phosphatidylcholines on TFo c-ring structure were examined. The DARR spectra at 15 ms mixing time are essentially similar to each other in saturated and unsaturated lipid membranes, suggesting that TFo c-rings have similar structures under the different environments. The spectrum of the sample in saturated lipid membranes showed better resolution and structural stability in the gel state. The C-terminal helix was suggested to locate in the outer layer of the c-ring.
Collapse
Affiliation(s)
- Su-Jin Kang
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, 565-0871, Japan
- Department of Biophysics and Chemical Biology, Seoul National University, Kwanak-Gu, Seoul, 151-742, Republic of Korea
| | - Yasuto Todokoro
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, 565-0871, Japan
| | - Suyeon Bak
- Department of Biophysics and Chemical Biology, Seoul National University, Kwanak-Gu, Seoul, 151-742, Republic of Korea
| | - Toshiharu Suzuki
- Department of Applied Chemistry, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Masasuke Yoshida
- Department of Molecular Bioscience, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto, 603-8555, Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, 565-0871, Japan
| | - Hideo Akutsu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, 565-0871, Japan.
- Department of Biophysics and Chemical Biology, Seoul National University, Kwanak-Gu, Seoul, 151-742, Republic of Korea.
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehirocho, Tsurumi-ku, Yokohama, 230-0045, Japan.
| |
Collapse
|
168
|
Oxidative Phosphorylation as a Target Space for Tuberculosis: Success, Caution, and Future Directions. Microbiol Spectr 2017; 5. [PMID: 28597820 DOI: 10.1128/microbiolspec.tbtb2-0014-2016] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The emergence and spread of drug-resistant pathogens, and our inability to develop new antimicrobials to combat resistance, have inspired scientists to seek out new targets for drug development. The Mycobacterium tuberculosis complex is a group of obligately aerobic bacteria that have specialized for inhabiting a wide range of intracellular and extracellular environments. Two fundamental features in this adaptation are the flexible utilization of energy sources and continued metabolism in the absence of growth. M. tuberculosis is an obligately aerobic heterotroph that depends on oxidative phosphorylation for growth and survival. However, several studies are redefining the metabolic breadth of the genus. Alternative electron donors and acceptors may provide the maintenance energy for the pathogen to maintain viability in hypoxic, nonreplicating states relevant to latent infection. This hidden metabolic flexibility may ultimately decrease the efficacy of drugs targeted against primary dehydrogenases and terminal oxidases. However, it may also open up opportunities to develop novel antimycobacterials targeting persister cells. In this review, we discuss the progress in understanding the role of energetic targets in mycobacterial physiology and pathogenesis and the opportunities for drug discovery.
Collapse
|
169
|
Li L, Jin Y, Wang B, Yang Z, Liu M, Guo H, Zhang J, Lu Y. A structure-based strategy toward the development of novel candidates for antimycobacterial activity: Synthesis, biological evaluation, and docking study. Chem Biol Drug Des 2017; 91:769-780. [DOI: 10.1111/cbdd.13142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/18/2017] [Accepted: 11/01/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Linhu Li
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yuanyuan Jin
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Bin Wang
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research; Department of Pharmacology; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Chest Hospital; Capital Medical University; Beijing China
| | - Zhaoyong Yang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Huiyuan Guo
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Jun Zhang
- Institute of Medicinal Biotechnology; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
- Zhejiang Starry Pharmaceutical Co. Ltd.; Xianju China
| | - Yu Lu
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research; Department of Pharmacology; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Chest Hospital; Capital Medical University; Beijing China
| |
Collapse
|
170
|
Fiorillo M, Lamb R, Tanowitz HB, Cappello AR, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Bedaquiline, an FDA-approved antibiotic, inhibits mitochondrial function and potently blocks the proliferative expansion of stem-like cancer cells (CSCs). Aging (Albany NY) 2017; 8:1593-607. [PMID: 27344270 PMCID: PMC5032685 DOI: 10.18632/aging.100983] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/06/2016] [Indexed: 12/31/2022]
Abstract
Bedaquiline (a.k.a., Sirturo) is an anti-microbial agent, which is approved by the FDA for the treatment of multi-drug resistant pulmonary tuberculosis (TB). Bedaquiline is a first-in-class diaryl-quinoline compound, that mechanistically inhibits the bacterial ATP-synthase, and shows potent activity against both drug-sensitive and drug-resistant TB. Interestingly, eukaryotic mitochondria originally evolved from engulfed aerobic bacteria. Thus, we hypothesized that, in mammalian cells, bedaquiline might also target the mitochondrial ATP-synthase, leading to mitochondrial dysfunction and ATP depletion. Here, we show that bedaquiline has anti-cancer activity, directed against Cancer Stem-like Cells (CSCs). More specifically, we demonstrate that bedaquiline treatment of MCF7 breast cancer cells inhibits mitochondrial oxygen-consumption, as well as glycolysis, but induces oxidative stress. Importantly, bedaquiline significantly blocks the propagation and expansion of MCF7-derived CSCs, with an IC-50 of approx. 1-μM, as determined using the mammosphere assay. Similarly, bedaquiline also reduces both the CD44+/CD24low/− CSC and ALDH+ CSC populations, under anchorage-independent growth conditions. In striking contrast, bedaquiline significantly increases oxygen consumption in normal human fibroblasts, consistent with the fact that it is well-tolerated in patients treated for TB infections. As such, future pre-clinical studies and human clinical trials in cancer patients may be warranted. Interestingly, we also highlight that bedaquiline shares certain structural similarities with trans-piceatannol and trans-resveratrol, which are known natural flavonoid inhibitors of the mitochondrial ATP-synthase (complex V) and show anti-aging properties.
Collapse
Affiliation(s)
- Marco Fiorillo
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.,The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.,The Department of Pharmacy, Health and Nutritional Sciences, The University of Calabria, Cosenza, Italy
| | - Rebecca Lamb
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.,The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| | - Herbert B Tanowitz
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Anna Rita Cappello
- The Department of Pharmacy, Health and Nutritional Sciences, The University of Calabria, Cosenza, Italy
| | | | - Federica Sotgia
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Michael P Lisanti
- The Breast Cancer Now Research Unit, Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK.,The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK
| |
Collapse
|
171
|
Bown L, Srivastava SK, Piercey BM, McIsaac CK, Tahlan K. Mycobacterial Membrane Proteins QcrB and AtpE: Roles in Energetics, Antibiotic Targets, and Associated Mechanisms of Resistance. J Membr Biol 2017; 251:105-117. [DOI: 10.1007/s00232-017-9997-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
|
172
|
Wu X, Li F, Wang X, Li C, Meng Q, Wang C, Huang J, Chen S, Zhu Z. Antibiotic bedaquiline effectively targets growth, survival and tumor angiogenesis of lung cancer through suppressing energy metabolism. Biochem Biophys Res Commun 2017; 495:267-272. [PMID: 29107691 DOI: 10.1016/j.bbrc.2017.10.136] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 10/25/2017] [Indexed: 12/26/2022]
Abstract
Tumor angiogenesis plays essential roles during lung cancer progression and metastasis. Therapeutic agent that targets both tumor cell and vascular endothelial cell may achieve additional anti-tumor efficacy. We demonstrate that bedaquiline, a FDA-approved antibiotic drug, effectively targets lung cancer cells and angiogenesis. Bedaquiline dose-dependently inhibits proliferation and induces apoptosis of a panel of lung cancer cell lines regardless of subtypes and molecular heterogeneity. Bedaquiline also inhibits capillary network formation of human lung tumor associated-endothelial cell (HLT-EC) on Matrigel and its multiple functions, such as spreading, proliferation and apoptosis, even in the presence of vascular endothelial growth factor (VEGF). We further demonstrate that bedaquiline acts on lung cancer cells and HLT-EC via inhibiting mitochondrial respiration and glycolysis, leading to ATP reduction and oxidative stress. Consistently, oxidative damage on DNA, protein and lipid were detected in cells exposed to bedaquiline. Importantly, the results obtained in in vitro cell culture are reproducible in in vivo xenograft lung cancer mouse model, confirming that bedaquiline suppresses lug tumor growth and angiogenesis, and increases oxidative stress. Our findings demonstrating that energy depletion is effectively against lung tumor cells and angiogenesis. Our work also provide pre-clinical evidence to repurpose antibiotic bedaquiline for lung cancer treatment.
Collapse
Affiliation(s)
- Xiaomu Wu
- Department of Neurology, The Central Hospital of Wuhan, Wuhan, People's Republic of China
| | - Fajiu Li
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Xiaojiang Wang
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Chenghong Li
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Qinghua Meng
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Chuanhai Wang
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Jie Huang
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Shi Chen
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China
| | - Ziyang Zhu
- Department of Respiratory Medicine, Wuhan No. 6 Hospital, Affiliated Hospital to Jianghan University, Wuhan, People's Republic of China.
| |
Collapse
|
173
|
Screening of antitubercular compound library identifies novel ATP synthase inhibitors of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2017. [PMID: 29523328 DOI: 10.1016/j.tube.2017.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A limited number of anti-tuberculosis drug candidates with novel mode of action have entered clinical trials in recent years. ATP synthase is one such validated drug target which has yielded a drug recently. The aim of this study was to identify the novel chemical scaffolds targeting the Mycobacterium tuberculosis (M. tuberculosis) ATP synthase. In this study, inverted membrane vesicles of Mycobacterium smegmatis were prepared to establish luciferin based ATP estimation assay. This assay was used to screen 700 compounds which were earlier found to be active on the whole cell of M. tuberculosis. Antibacterial activity of hits against various susceptible and drug-resistant strains of M. tuberculosis was evaluated using the microplate alamar blue assay and their cytotoxicity was also determined to select the safe compounds for further study. Screening of 700 compounds resulted in the identification of two compounds (5228485 and 5220632) exhibiting an IC50 of 0.32 and 4.0 μg/ml respectively. Both compounds showed excellent anti-TB activity (MIC of 0.5-2.0 μg/ml against Mtb H37Rv) and low cytotoxicity in human cell line and sub-mitochondrial particles. The three-dimensional structure of M. tuberculosis ATPase was predicted using in-silico approach and docking studies were performed with the active compounds. The interaction between compounds and bacterial ATP synthase was confirmed by molecular docking analysis. In conclusion screening of compound library has resulted in the identification of two novel chemical scaffolds targeting mycobacterial ATP synthase.
Collapse
|
174
|
Bedaquiline Inhibits the ATP Synthase in Mycobacterium abscessus and Is Effective in Infected Zebrafish. Antimicrob Agents Chemother 2017; 61:AAC.01225-17. [PMID: 28807917 DOI: 10.1128/aac.01225-17] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/08/2017] [Indexed: 12/24/2022] Open
Abstract
Pulmonary infections caused by Mycobacterium abscessus are emerging as a global threat, especially in cystic fibrosis patients. Further intensifying the concern of M. abscessus infection is the recent evidence of human-to-human transmission of the infection. M. abscessus is a naturally multidrug-resistant fast-growing pathogen for which pharmacological options are limited. Repurposing antitubercular drugs represents an attractive option for the development of chemotherapeutic alternatives against M. abscessus infections. Bedaquiline (BDQ), an ATP synthase inhibitor, has recently been approved for the treatment of multidrug-resistant tuberculosis. Herein, we show that BDQ has a very low MIC against a vast panel of clinical isolates. Despite being bacteriostatic in vitro, BDQ was highly efficacious in a zebrafish model of M. abscessus infection. Remarkably, a very short period of treatment was sufficient to protect the infected larvae from M. abscessus-induced killing. This was corroborated with reduced numbers of abscesses and cords, considered to be major pathophysiological signs in infected zebrafish. Mode-of-action studies revealed that BDQ triggered a rapid depletion of ATP in M. abscessusin vitro, consistent with the drug targeting the FoF1 ATP synthase. Importantly, despite a failure to select in vitro for spontaneous mutants that are highly resistant to BDQ, the transfer of single nucleotide polymorphisms leading to D29V or A64P substitutions in atpE conferred high resistance, thus resolving the target of BDQ in M. abscessus Overall, this study indicates that BDQ is active against M. abscessusin vitro and in vivo and should be considered for clinical use against the difficult-to-manage M. abscessus pulmonary infections.
Collapse
|
175
|
Tong AST, Choi PJ, Blaser A, Sutherland HS, Tsang SKY, Guillemont J, Motte M, Cooper CB, Andries K, Van den Broeck W, Franzblau SG, Upton AM, Denny WA, Palmer BD, Conole D. 6-Cyano Analogues of Bedaquiline as Less Lipophilic and Potentially Safer Diarylquinolines for Tuberculosis. ACS Med Chem Lett 2017; 8:1019-1024. [PMID: 29057044 PMCID: PMC5642017 DOI: 10.1021/acsmedchemlett.7b00196] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/22/2017] [Indexed: 12/26/2022] Open
Abstract
![]()
Bedaquiline (1) is a
new drug for tuberculosis and the first of the diarylquinoline class.
It demonstrates excellent efficacy against TB but induces phospholipidosis
at high doses, has a long terminal elimination half-life (due to its
high lipophilicity), and exhibits potent hERG channel inhibition,
resulting in clinical QTc interval prolongation. A number of structural
ring A analogues of bedaquiline have been prepared and evaluated for
their anti-M.tb activity (MIC90), with
a view to their possible application as less lipophilic second generation
compounds. It was previously observed that a range of 6-substituted
analogues of 1 demonstrated a positive correlation between
potency (MIC90) toward M.tb and drug lipophilicity.
Contrary to this trend, we discovered, by virtue of a clogP/M.tb score, that a 6-cyano (CN) substituent provides a substantial
reduction in lipophilicity with only modest effects on MIC values,
suggesting this substituent as a useful tool in the search for effective
and safer analogues of 1.
Collapse
Affiliation(s)
- Amy S. T. Tong
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Peter J. Choi
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Adrian Blaser
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Hamish S. Sutherland
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Sophia K. Y. Tsang
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jerome Guillemont
- Medicinal Chemistry
Department (Infectious Diseases), Janssen Pharmaceuticals, Campus
de Maigremont, BP315, 27106 Val de Reuil Cedex, France
| | - Magali Motte
- Medicinal Chemistry
Department (Infectious Diseases), Janssen Pharmaceuticals, Campus
de Maigremont, BP315, 27106 Val de Reuil Cedex, France
| | - Christopher B. Cooper
- Global Alliance for TB Drug Development, 40 Wall Street, New York, New York 10005, United States
| | - Koen Andries
- Infectious Diseases BVBA, Janssen Pharmaceuticals, Beerse, Belgium
| | | | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Anna M. Upton
- Global Alliance for TB Drug Development, 40 Wall Street, New York, New York 10005, United States
| | - William A. Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Brian D. Palmer
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Daniel Conole
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
176
|
Nesci S, Trombetti F, Ventrella V, Pagliarani A. Post-translational modifications of the mitochondrial F 1F O-ATPase. Biochim Biophys Acta Gen Subj 2017; 1861:2902-2912. [PMID: 28782624 DOI: 10.1016/j.bbagen.2017.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The mitochondrial F1FO-ATPase has the main role in synthesizing most of ATP, thus providing energy to living cells, but it also works in reverse and hydrolyzes ATP, depending on the transmembrane electrochemical gradient. Within the same complex the vital role of the enzyme of life coexists with that of molecular switch to trigger programmed cell death. The two-faced vital/lethal role makes the enzyme complex an intriguing biochemical target to fight pathogens resistant to traditional therapies and diseases linked to mitochondrial dysfunctions. A variety of post-translational modifications (PTMs) of selected F1FO-ATPase aminoacids have been reported to affect the enzyme function. SCOPE OF REVIEW By reviewing the known PTMs of aminoacid side chains of both F1 and FO sectors according to the most recent advances, the main aim is to highlight how local chemical changes may constitute the molecular key leading to pathological or physiological events. MAJOR CONCLUSIONS PTMs represent the chemical tool to modulate the F1FO-ATPase activity in response to different stimuli. Some PTMs are required to ensure the enzyme catalysis or, conversely, to inactivate the enzyme function. Each covalent modification of the F1FO-ATPase, which occur in response to local changes, is the result of a selective molecular mechanism which, by translating a chemical modification into a biochemical effect, guarantees the enzyme tuning under changing conditions. GENERAL SIGNIFICANCE Once highlighted how the molecular mechanism works, some PTMs may be exploited to modulate the effect of drugs targeting the enzyme complex or constitute promising tools for F1FO-ATPase-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Salvatore Nesci
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, via Tolara di Sopra 50, 40064 Ozzano dell'Emilia, BO, Italy
| | - Fabiana Trombetti
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, via Tolara di Sopra 50, 40064 Ozzano dell'Emilia, BO, Italy
| | - Vittoria Ventrella
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, via Tolara di Sopra 50, 40064 Ozzano dell'Emilia, BO, Italy
| | - Alessandra Pagliarani
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, via Tolara di Sopra 50, 40064 Ozzano dell'Emilia, BO, Italy.
| |
Collapse
|
177
|
Ragunathan P, Sielaff H, Sundararaman L, Biuković G, Subramanian Manimekalai MS, Singh D, Kundu S, Wohland T, Frasch W, Dick T, Grüber G. The uniqueness of subunit α of mycobacterial F-ATP synthases: An evolutionary variant for niche adaptation. J Biol Chem 2017; 292:11262-11279. [PMID: 28495884 PMCID: PMC5500794 DOI: 10.1074/jbc.m117.784959] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/11/2017] [Indexed: 01/30/2023] Open
Abstract
The F1F0 -ATP (F-ATP) synthase is essential for growth of Mycobacterium tuberculosis, the causative agent of tuberculosis (TB). In addition to their synthase function most F-ATP synthases possess an ATP-hydrolase activity, which is coupled to proton-pumping activity. However, the mycobacterial enzyme lacks this reverse activity, but the reason for this deficiency is unclear. Here, we report that a Mycobacterium-specific, 36-amino acid long C-terminal domain in the nucleotide-binding subunit α (Mtα) of F-ATP synthase suppresses its ATPase activity and determined the mechanism of suppression. First, we employed vesicles to show that in intact membrane-embedded mycobacterial F-ATP synthases deletion of the C-terminal domain enabled ATPase and proton-pumping activity. We then generated a heterologous F-ATP synthase model system, which demonstrated that transfer of the mycobacterial C-terminal domain to a standard F-ATP synthase α subunit suppresses ATPase activity. Single-molecule rotation assays indicated that the introduction of this Mycobacterium-specific domain decreased the angular velocity of the power-stroke after ATP binding. Solution X-ray scattering data and NMR results revealed the solution shape of Mtα and the 3D structure of the subunit α C-terminal peptide 521PDEHVEALDEDKLAKEAVKV540 of M. tubercolosis (Mtα(521-540)), respectively. Together with cross-linking studies, the solution structural data lead to a model, in which Mtα(521-540) comes in close proximity with subunit γ residues 104-109, whose interaction may influence the rotation of the camshaft-like subunit γ. Finally, we propose that the unique segment Mtα(514-549), which is accessible at the C terminus of mycobacterial subunit α, is a promising drug epitope.
Collapse
Affiliation(s)
- Priya Ragunathan
- From the Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Hendrik Sielaff
- From the Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Lavanya Sundararaman
- From the Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Goran Biuković
- the Department of Microbiology and Immunology, National University of Singapore, Yong Loo Lin School of Medicine, 14 Medical Drive, Singapore 117599, Republic of Singapore
| | | | - Dhirendra Singh
- From the Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Subhashri Kundu
- the Department of Microbiology and Immunology, National University of Singapore, Yong Loo Lin School of Medicine, 14 Medical Drive, Singapore 117599, Republic of Singapore
| | - Thorsten Wohland
- the Departments of Biological Sciences and Chemistry and NUS Centre for Bioimaging Sciences (CBIS), National University of Singapore, 14 Science Drive 4, Singapore 117557, Republic of Singapore, and
| | - Wayne Frasch
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287
| | - Thomas Dick
- the Department of Microbiology and Immunology, National University of Singapore, Yong Loo Lin School of Medicine, 14 Medical Drive, Singapore 117599, Republic of Singapore
| | - Gerhard Grüber
- From the Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Republic of Singapore,
| |
Collapse
|
178
|
Antimicrobial Activity of Quinazolin Derivatives of 1,2-Di(quinazolin-4-yl)diselane against Mycobacteria. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5791781. [PMID: 28612027 PMCID: PMC5458374 DOI: 10.1155/2017/5791781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/12/2017] [Accepted: 04/03/2017] [Indexed: 11/17/2022]
Abstract
Mycobacterium tuberculosis (M. tuberculosis) is one of the leading causes of morbidity and mortality. Currently, the emergence of drug resistance has an urgent need for new drugs. In previous study, we found that 1,2-di(quinazolin-4-yl)diselane (DQYD), a quinazoline derivative, has anticancer activities against many cancers. However, whether DQYD has the activity of antimycobacterium is still little known. Here our results show that DQYD has a similar value of the minimum inhibitory concentration with clinical drugs against mycobacteria and also has the ability of bacteriostatic activity with dose-dependent and time-dependent manner. Furthermore, the activities of DQYD against M. tuberculosis are associated with intracellular ATP homeostasis. Meanwhile, mycobacterium DNA damage level was increased after DQYD treatment. But there was no correlation between survival of mycobacteria in the presence of DQYD and intercellular reactive oxygen species. This study enlightens the possible benefits of quinazoline derivatives as potential antimycobacterium compounds and furtherly suggests a new strategy to develop new methods for searching antituberculosis drugs.
Collapse
|
179
|
AlMatar M, AlMandeal H, Var I, Kayar B, Köksal F. New drugs for the treatment of Mycobacterium tuberculosis infection. Biomed Pharmacother 2017; 91:546-558. [PMID: 28482292 DOI: 10.1016/j.biopha.2017.04.105] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/29/2017] [Accepted: 04/23/2017] [Indexed: 12/25/2022] Open
Abstract
Tuberculosis presents a grave challenge to health, globally instigating 1.5 million mortalities each year. Following the breakthrough of first-line anti-TB medication, the number of mortalities reduced greatly; nonetheless, the swift appearance of tuberculosis which was drug-resistant, as well as the capability of the bacterium to survive and stay dormant are a considerable problem for public health. In order to address this issue, several novel possible candidates for tuberculosis therapy have been subjected to clinical trials of late. The novel antimycobacterial agents are acquired from different categories of medications, operate through a range of action systems, and are at various phases of advancement. We therefore talk about the present methods of treating tuberculosis and novel anti-TB agents with their action method, in order to advance awareness of these new compounds and medications.
Collapse
Affiliation(s)
- Manaf AlMatar
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitüsü), Cukurova University, Adana, Turkey.
| | - Husam AlMandeal
- Universitätsklinikum des Saarlandes, Gebäude 90, Kirrberger Straße, D-66421, Homburg, Germany
| | - Işıl Var
- Department of Food Engineering, Agricultural Faculty, Cukurova University, Adana, Turkey
| | - Begüm Kayar
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
180
|
Targeting Energy Metabolism in Mycobacterium tuberculosis, a New Paradigm in Antimycobacterial Drug Discovery. mBio 2017; 8:mBio.00272-17. [PMID: 28400527 PMCID: PMC5388804 DOI: 10.1128/mbio.00272-17] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Drug-resistant mycobacterial infections are a serious global health challenge, leading to high mortality and socioeconomic burdens in developing countries worldwide. New innovative approaches, from identification of new targets to discovery of novel chemical scaffolds, are urgently needed. Recently, energy metabolism in mycobacteria, in particular the oxidative phosphorylation pathway, has emerged as an object of intense microbiological investigation and as a novel target pathway in drug discovery. New classes of antibacterials interfering with elements of the oxidative phosphorylation pathway are highly active in combating dormant or latent mycobacterial infections, with a promise of shortening tuberculosis chemotherapy. The regulatory approval of the ATP synthase inhibitor bedaquiline and the discovery of Q203, a candidate drug targeting the cytochrome bc1 complex, have highlighted the central importance of this new target pathway. In this review, we discuss key features and potential applications of inhibiting energy metabolism in our quest for discovering potent novel and sterilizing drug combinations for combating tuberculosis. We believe that the combination of drugs targeting elements of the oxidative phosphorylation pathway can lead to a completely new regimen for drug-susceptible and multidrug-resistant tuberculosis.
Collapse
|
181
|
Schulz S, Wilkes M, Mills DJ, Kühlbrandt W, Meier T. Molecular architecture of the N-type ATPase rotor ring from Burkholderia pseudomallei. EMBO Rep 2017; 18:526-535. [PMID: 28283532 PMCID: PMC5376962 DOI: 10.15252/embr.201643374] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/02/2017] [Accepted: 02/09/2017] [Indexed: 11/09/2022] Open
Abstract
The genome of the highly infectious bacterium Burkholderia pseudomallei harbors an atp operon that encodes an N‐type rotary ATPase, in addition to an operon for a regular F‐type rotary ATPase. The molecular architecture of N‐type ATPases is unknown and their biochemical properties and cellular functions are largely unexplored. We studied the B. pseudomallei N1No‐type ATPase and investigated the structure and ion specificity of its membrane‐embedded c‐ring rotor by single‐particle electron cryo‐microscopy. Of several amphiphilic compounds tested for solubilizing the complex, the choice of the low‐density, low‐CMC detergent LDAO was optimal in terms of map quality and resolution. The cryoEM map of the c‐ring at 6.1 Å resolution reveals a heptadecameric oligomer with a molecular mass of ~141 kDa. Biochemical measurements indicate that the c17 ring is H+ specific, demonstrating that the ATPase is proton‐coupled. The c17 ring stoichiometry results in a very high ion‐to‐ATP ratio of 5.7. We propose that this N‐ATPase is a highly efficient proton pump that helps these melioidosis‐causing bacteria to survive in the hostile, acidic environment of phagosomes.
Collapse
Affiliation(s)
- Sarah Schulz
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Martin Wilkes
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Deryck J Mills
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Werner Kühlbrandt
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Thomas Meier
- Department of Structural Biology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| |
Collapse
|
182
|
Tantry SJ, Markad SD, Shinde V, Bhat J, Balakrishnan G, Gupta AK, Ambady A, Raichurkar A, Kedari C, Sharma S, Mudugal NV, Narayan A, Naveen Kumar CN, Nanduri R, Bharath S, Reddy J, Panduga V, Prabhakar KR, Kandaswamy K, Saralaya R, Kaur P, Dinesh N, Guptha S, Rich K, Murray D, Plant H, Preston M, Ashton H, Plant D, Walsh J, Alcock P, Naylor K, Collier M, Whiteaker J, McLaughlin RE, Mallya M, Panda M, Rudrapatna S, Ramachandran V, Shandil R, Sambandamurthy VK, Mdluli K, Cooper CB, Rubin H, Yano T, Iyer P, Narayanan S, Kavanagh S, Mukherjee K, Balasubramanian V, Hosagrahara VP, Solapure S, Ravishankar S, Hameed P S. Discovery of Imidazo[1,2-a]pyridine Ethers and Squaramides as Selective and Potent Inhibitors of Mycobacterial Adenosine Triphosphate (ATP) Synthesis. J Med Chem 2017; 60:1379-1399. [PMID: 28075132 DOI: 10.1021/acs.jmedchem.6b01358] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The approval of bedaquiline to treat tuberculosis has validated adenosine triphosphate (ATP) synthase as an attractive target to kill Mycobacterium tuberculosis (Mtb). Herein, we report the discovery of two diverse lead series imidazo[1,2-a]pyridine ethers (IPE) and squaramides (SQA) as inhibitors of mycobacterial ATP synthesis. Through medicinal chemistry exploration, we established a robust structure-activity relationship of these two scaffolds, resulting in nanomolar potencies in an ATP synthesis inhibition assay. A biochemical deconvolution cascade suggested cytochrome c oxidase as the potential target of IPE class of molecules, whereas characterization of spontaneous resistant mutants of SQAs unambiguously identified ATP synthase as its molecular target. Absence of cross resistance against bedaquiline resistant mutants suggested a different binding site for SQAs on ATP synthase. Furthermore, SQAs were found to be noncytotoxic and demonstrated efficacy in a mouse model of tuberculosis infection.
Collapse
Affiliation(s)
- Subramanyam J Tantry
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Shankar D Markad
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Vikas Shinde
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Jyothi Bhat
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Gayathri Balakrishnan
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Amit K Gupta
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Anisha Ambady
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Anandkumar Raichurkar
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Chaitanyakumar Kedari
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Sreevalli Sharma
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Naina V Mudugal
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Ashwini Narayan
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - C N Naveen Kumar
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Robert Nanduri
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Sowmya Bharath
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Jitendar Reddy
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Vijender Panduga
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - K R Prabhakar
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Karthikeyan Kandaswamy
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Ramanatha Saralaya
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Parvinder Kaur
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Neela Dinesh
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Supreeth Guptha
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Kirsty Rich
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - David Murray
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Helen Plant
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Marian Preston
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Helen Ashton
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Darren Plant
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Jarrod Walsh
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Peter Alcock
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Kathryn Naylor
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Matthew Collier
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - James Whiteaker
- Infection Innovative Medicines, AstraZeneca , 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Robert E McLaughlin
- Infection Innovative Medicines, AstraZeneca , 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Meenakshi Mallya
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Manoranjan Panda
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Suresh Rudrapatna
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Vasanthi Ramachandran
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Radha Shandil
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Vasan K Sambandamurthy
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Khisi Mdluli
- Global Alliance for TB Drug Development , 40 Wall Street, 24th Floor, New York, New York 10005, United States
| | - Christopher B Cooper
- Global Alliance for TB Drug Development , 40 Wall Street, 24th Floor, New York, New York 10005, United States
| | - Harvey Rubin
- University of Pennsylvania , 111 Clinical Research Building, 415 Curie Boulevard, Philadelphia Pennsylvania 19104, United States
| | - Takahiro Yano
- University of Pennsylvania , 111 Clinical Research Building, 415 Curie Boulevard, Philadelphia Pennsylvania 19104, United States
| | - Pravin Iyer
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Shridhar Narayanan
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Stefan Kavanagh
- AstraZeneca , Alderley Park, Mereside, Macclesfield, Cheshire U.K. SK10 4TG
| | - Kakoli Mukherjee
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - V Balasubramanian
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Vinayak P Hosagrahara
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Suresh Solapure
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Sudha Ravishankar
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| | - Shahul Hameed P
- Innovative Medicines, AstraZeneca India Pvt. Ltd. , Bellary Road, Hebbal, Bangalore 560024, India
| |
Collapse
|
183
|
Analysis of an N-terminal deletion in subunit a of the Escherichia coli ATP synthase. J Bioenerg Biomembr 2017; 49:171-181. [PMID: 28078625 DOI: 10.1007/s10863-017-9694-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
Abstract
Subunit a is a membrane-bound stator subunit of the ATP synthase and is essential for proton translocation. The N-terminus of subunit a in E. coli is localized to the periplasm, and contains a sequence motif that is conserved among some bacteria. Previous work has identified mutations in this region that impair enzyme activity. Here, an internal deletion was constructed in subunit a in which residues 6-20 were replaced by a single lysine residue, and this mutant was unable to grow on succinate minimal medium. Membrane vesicles prepared from this mutant lacked ATP synthesis and ATP-driven proton translocation, even though immunoblots showed a significant level of subunit a. Similar results were obtained after purification and reconstitution of the mutant ATP synthase into liposomes. The location of subunit a with respect to its neighboring subunits b and c was probed by introducing cysteine substitutions that were known to promote cross-linking: a_L207C + c_I55C, a_L121C + b_N4C, and a_T107C + b_V18C. The last pair was unable to form cross-links in the background of the deletion mutant. The results indicate that loss of the N-terminal region of subunit a does not generally disrupt its structure, but does alter interactions with subunit b.
Collapse
|
184
|
Hartman TE, Wang Z, Jansen RS, Gardete S, Rhee KY. Metabolic Perspectives on Persistence. Microbiol Spectr 2017; 5:10.1128/microbiolspec.TBTB2-0026-2016. [PMID: 28155811 PMCID: PMC5302851 DOI: 10.1128/microbiolspec.tbtb2-0026-2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence has left little doubt about the importance of persistence or metabolism in the biology and chemotherapy of tuberculosis. However, knowledge of the intersection between these two factors has only recently begun to emerge. Here, we provide a focused review of metabolic characteristics associated with Mycobacterium tuberculosis persistence. We focus on metabolism because it is the biochemical foundation of all physiologic processes and a distinguishing hallmark of M. tuberculosis physiology and pathogenicity. In addition, it serves as the chemical interface between host and pathogen. Existing knowledge, however, derives largely from physiologic contexts in which replication is the primary biochemical objective. The goal of this review is to reframe current knowledge of M. tuberculosis metabolism in the context of persistence, where quiescence is often a key distinguishing characteristic. Such a perspective may help ongoing efforts to develop more efficient cures and inform on novel strategies to break the cycle of transmission sustaining the pandemic.
Collapse
Affiliation(s)
- Travis E. Hartman
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
| | - Zhe Wang
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
| | - Robert S. Jansen
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
| | - Susana Gardete
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
| | - Kyu Y. Rhee
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
- Department of Microbiology & Immunology, Division of Infectious Diseases, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
185
|
Emergence of mmpT5 Variants during Bedaquiline Treatment of Mycobacterium intracellulare Lung Disease. J Clin Microbiol 2016; 55:574-584. [PMID: 27927925 DOI: 10.1128/jcm.02087-16] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/01/2016] [Indexed: 11/20/2022] Open
Abstract
Bedaquiline (BDQ), a diarylquinoline antibiotic that targets ATP synthase, is effective for the treatment of Mycobacterium tuberculosis infections that no longer respond to conventional drugs. While investigating the off-label use of BDQ as salvage therapy, seven of 13 patients with Mycobacterium intracellulare lung disease had an initial microbiological response and then relapsed. Whole-genome comparison of pretreatment and relapse isolates of M. intracellulare uncovered mutations in a previously uncharacterized locus, mmpT5 Preliminary analysis suggested similarities between mmpT5 and the mmpR5 locus, which is associated with low-level BDQ resistance in M. tuberculosis Both genes encode transcriptional regulators and are adjacent to orthologs of the mmpS5-mmpL5 drug efflux operon. However, MmpT5 belongs to the TetR superfamily, whereas MmpR5 is a MarR family protein. Targeted sequencing uncovered nonsynonymous mmpT5 mutations in isolates from all seven relapse cases, including two pretreatment isolates. In contrast, only two relapse patient isolates had nonsynonymous changes in ATP synthase subunit c (atpE), the primary target of BDQ. Susceptibility testing indicated that mmpT5 mutations are associated with modest 2- to 8-fold increases in MICs for BDQ and clofazimine, whereas one atpE mutant exhibited a 50-fold increase in MIC for BDQ. Bedaquiline shows potential for the treatment of M. intracellulare lung disease, but optimization of treatment regimens is required to prevent the emergence of mmpT5 variants and microbiological relapse.
Collapse
|
186
|
Leone V, Faraldo-Gómez JD. Structure and mechanism of the ATP synthase membrane motor inferred from quantitative integrative modeling. J Gen Physiol 2016; 148:441-457. [PMID: 27821609 PMCID: PMC5129741 DOI: 10.1085/jgp.201611679] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/13/2016] [Indexed: 01/31/2023] Open
Abstract
The ATP synthase is a molecular rotor that recycles ADP into ATP. Leone and Faraldo-Gómez use structural modeling to reinterpret and reconcile recent cryo-EM data for its membrane domain with other experimental evidence, gaining insights into its mechanism and the mode of inhibition by oligomycin. Two subunits within the transmembrane domain of the ATP synthase—the c-ring and subunit a—energize the production of 90% of cellular ATP by transducing an electrochemical gradient of H+ or Na+ into rotational motion. The nature of this turbine-like energy conversion mechanism has been elusive for decades, owing to the lack of definitive structural information on subunit a or its c-ring interface. In a recent breakthrough, several structures of this complex were resolved by cryo–electron microscopy (cryo-EM), but the modest resolution of the data has led to divergent interpretations. Moreover, the unexpected architecture of the complex has cast doubts on a wealth of earlier biochemical analyses conducted to probe this structure. Here, we use quantitative molecular-modeling methods to derive a structure of the a–c complex that is not only objectively consistent with the cryo-EM data, but also with correlated mutation analyses of both subunits and with prior cross-linking and cysteine accessibility measurements. This systematic, integrative approach reveals unambiguously the topology of subunit a and its relationship with the c-ring. Mapping of known Cd2+ block sites and conserved protonatable residues onto the structure delineates two noncontiguous pathways across the complex, connecting two adjacent proton-binding sites in the c-ring to the space on either side of the membrane. The location of these binding sites and of a strictly conserved arginine on subunit a, which serves to prevent protons from hopping between them, explains the directionality of the rotary mechanism and its strict coupling to the proton-motive force. Additionally, mapping of mutations conferring resistance to oligomycin unexpectedly reveals that this prototypical inhibitor may bind to two distinct sites at the a–c interface, explaining its ability to block the mechanism of the enzyme irrespective of the direction of rotation of the c-ring. In summary, this study is a stepping stone toward establishing the mechanism of the ATP synthase at the atomic level.
Collapse
Affiliation(s)
- Vanessa Leone
- Theoretical Molecular Biophysics Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - José D Faraldo-Gómez
- Theoretical Molecular Biophysics Section, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
187
|
He C, Preiss L, Wang B, Fu L, Wen H, Zhang X, Cui H, Meier T, Yin D. Structural Simplification of Bedaquiline: the Discovery of 3-(4-(N,N-Dimethylaminomethyl)phenyl)quinoline-Derived Antitubercular Lead Compounds. ChemMedChem 2016; 12:106-119. [PMID: 27792278 PMCID: PMC5298006 DOI: 10.1002/cmdc.201600441] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Indexed: 01/03/2023]
Abstract
Bedaquiline (BDQ) is a novel and highly potent last-line antituberculosis drug that was approved by the US FDA in 2013. Owing to its stereo-structural complexity, chemical synthesis and compound optimization are rather difficult and expensive. This study describes the structural simplification of bedaquiline while preserving antitubercular activity. The compound's structure was split into fragments and reassembled in various combinations while replacing the two chiral carbon atoms with an achiral linkage instead. Four series of analogues were designed; these candidates retained their potent antitubercular activity at sub-microgram per mL concentrations against both sensitive and multidrug-resistant (MDR) Mycobacterium tuberculosis strains. Six out of the top nine MIC-ranked candidates were found to inhibit mycobacterial ATP synthesis activity with IC50 values between 20 and 40 μm, one had IC50 >66 μm, and two showed no inhibition, despite their antitubercular activity. These results provide a basis for the development of chemically less complex, lower-cost bedaquiline derivatives and describe the identification of two derivatives with antitubercular activity against non-ATP synthase related targets.
Collapse
Affiliation(s)
- Chunxian He
- State Key Laboratory of Bioactive Substances and Function ofNatural Medicine, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China.,Beijing Key Laboratory of Active Substances Discovery and DrugabilityEvaluation, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China
| | - Laura Preiss
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany
| | - Bin Wang
- Department of Pharmacology, Beijing Tuberculosis and Thoracic TumorResearch Institute, Beijing Chest Hospital, Capital Medical University, 97 Ma Chang Street, Beijing, 101149, China
| | - Lei Fu
- Department of Pharmacology, Beijing Tuberculosis and Thoracic TumorResearch Institute, Beijing Chest Hospital, Capital Medical University, 97 Ma Chang Street, Beijing, 101149, China
| | - Hui Wen
- Beijing Key Laboratory of Active Substances Discovery and DrugabilityEvaluation, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China
| | - Xiang Zhang
- Beijing Key Laboratory of Active Substances Discovery and DrugabilityEvaluation, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China
| | - Huaqing Cui
- Beijing Key Laboratory of Active Substances Discovery and DrugabilityEvaluation, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China
| | - Thomas Meier
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany.,Department of Life Sciences, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Function ofNatural Medicine, Institute of Materia Medica, Peking Union Medical College andChinese Academy of Medical Sciences, Beijing, 100050, China
| |
Collapse
|
188
|
Zhou W, Leone V, Krah A, Faraldo-Gómez JD. Predicted Structures of the Proton-Bound Membrane-Embedded Rotor Rings of the Saccharomyces cerevisiae and Escherichia coli ATP Synthases. J Phys Chem B 2016; 121:3297-3307. [PMID: 27715045 DOI: 10.1021/acs.jpcb.6b08051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent years have witnessed a renewed interest in the ATP synthase as a drug target against human pathogens. Indeed, clinical, biochemical, and structural data indicate that hydrophobic inhibitors targeting the membrane-embedded proton-binding sites of the c-subunit ring could serve as last-resort antibiotics against multidrug resistant strains. However, because inhibition of the mitochondrial ATP synthase in humans is lethal, it is essential that these inhibitors be not only potent but also highly selective for the bacterial enzyme. To this end, a detailed understanding of the structure of this protein target is arguably instrumental. Here, we use computational methods to predict the atomic structures of the proton-binding sites in two prototypical c-rings: that of the ATP synthase from Saccharomyces cerevisiae, which is a model system for mitochondrial enzymes, and that from Escherichia coli, which can be pathogenic for humans. Our study reveals the structure of these binding sites loaded with protons and in the context of the membrane, that is, in the state that would mediate the recognition of a potential inhibitor. Both structures reflect a mode of proton coordination unlike those previously observed in other c-ring structures, whether experimental or modeled.
Collapse
Affiliation(s)
- Wenchang Zhou
- Theoretical Molecular Biophysics Section, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Vanessa Leone
- Theoretical Molecular Biophysics Section, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Alexander Krah
- Theoretical Molecular Biophysics Group, Max Planck Institute of Biophysics , 60438 Frankfurt am Main, Germany
| | - José D Faraldo-Gómez
- Theoretical Molecular Biophysics Section, National Heart, Lung and Blood Institute, National Institutes of Health , Bethesda, Maryland 20892, United States.,Theoretical Molecular Biophysics Group, Max Planck Institute of Biophysics , 60438 Frankfurt am Main, Germany
| |
Collapse
|
189
|
Bedaquiline Targets the ε Subunit of Mycobacterial F-ATP Synthase. Antimicrob Agents Chemother 2016; 60:6977-6979. [PMID: 27620476 PMCID: PMC5075122 DOI: 10.1128/aac.01291-16] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/04/2016] [Indexed: 12/24/2022] Open
Abstract
The tuberculosis drug bedaquiline inhibits mycobacterial F-ATP synthase by binding to its c subunit. Using the purified ε subunit of the synthase and spectroscopy, we previously demonstrated that the drug interacts with this protein near its unique tryptophan residue. Here, we show that replacement of ε's tryptophan with alanine resulted in bedaquiline hypersusceptibility of the bacteria. Overexpression of the wild-type ε subunit caused resistance. These results suggest that the drug also targets the ε subunit.
Collapse
|
190
|
Cholo MC, Mothiba MT, Fourie B, Anderson R. Mechanisms of action and therapeutic efficacies of the lipophilic antimycobacterial agents clofazimine and bedaquiline. J Antimicrob Chemother 2016; 72:338-353. [PMID: 27798208 DOI: 10.1093/jac/dkw426] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Drug-resistant (DR)-TB is the major challenge confronting the global TB control programme, necessitating treatment with second-line anti-TB drugs, often with limited therapeutic efficacy. This scenario has resulted in the inclusion of Group 5 antibiotics in various therapeutic regimens, two of which promise to impact significantly on the outcome of the therapy of DR-TB. These are the 're-purposed' riminophenazine, clofazimine, and the recently approved diarylquinoline, bedaquiline. Although they differ structurally, both of these lipophilic agents possess cationic amphiphilic properties that enable them to target and inactivate essential ion transporters in the outer membrane of Mycobacterium tuberculosis. In the case of bedaquiline, the primary target is the key respiratory chain enzyme F1/F0-ATPase, whereas clofazimine is less selective, apparently inhibiting several targets, which may underpin the extremely low level of resistance to this agent. This review is focused on similarities and differences between clofazimine and bedaquiline, specifically in respect of molecular mechanisms of antimycobacterial action, targeting of quiescent and metabolically active organisms, therapeutic efficacy in the clinical setting of DR-TB, resistance mechanisms, pharmacodynamics, pharmacokinetics and adverse events.
Collapse
Affiliation(s)
- Moloko C Cholo
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Maborwa T Mothiba
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Bernard Fourie
- Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Ronald Anderson
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| |
Collapse
|
191
|
Lamprecht DA, Finin PM, Rahman MA, Cumming BM, Russell SL, Jonnala SR, Adamson JH, Steyn AJC. Turning the respiratory flexibility of Mycobacterium tuberculosis against itself. Nat Commun 2016; 7:12393. [PMID: 27506290 PMCID: PMC4987515 DOI: 10.1038/ncomms12393] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 06/28/2016] [Indexed: 11/21/2022] Open
Abstract
The Mycobacterium tuberculosis (Mtb) electron transport chain (ETC) has received significant attention as a drug target, however its vulnerability may be affected by its flexibility in response to disruption. Here we determine the effect of the ETC inhibitors bedaquiline, Q203 and clofazimine on the Mtb ETC, and the value of the ETC as a drug target, by measuring Mtb's respiration using extracellular flux technology. We find that Mtb's ETC rapidly reroutes around inhibition by these drugs and increases total respiration to maintain ATP levels. Rerouting is possible because Mtb rapidly switches between terminal oxidases, and, unlike eukaryotes, is not susceptible to back pressure. Increased ETC activity potentiates clofazimine's production of reactive oxygen species, causing rapid killing in vitro and in a macrophage model. Our results indicate that combination therapy targeting the ETC can be exploited to enhance killing of Mtb.
Collapse
Affiliation(s)
- Dirk A. Lamprecht
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Peter M. Finin
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
- Department of Internal Medicine, University of Pittsburgh, 1218 Scaife Hall 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, USA
| | - Md. Aejazur Rahman
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Bridgette M. Cumming
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Shannon L. Russell
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | | | - John H. Adamson
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Adrie J. C. Steyn
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
- Department of Microbiology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, Alabama 35294-2170, USA
- Centres for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, Alabama 35294-2170, USA
| |
Collapse
|
192
|
Cardiolipin binds selectively but transiently to conserved lysine residues in the rotor of metazoan ATP synthases. Proc Natl Acad Sci U S A 2016; 113:8687-92. [PMID: 27382158 PMCID: PMC4978264 DOI: 10.1073/pnas.1608396113] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The anionic lipid cardiolipin is an essential component of active ATP synthases. In metazoans, their rotors contain a ring of eight c-subunits consisting of inner and outer circles of N- and C-terminal α-helices, respectively. The beginning of the C-terminal α-helix contains a strictly conserved and fully trimethylated lysine residue in the lipid head-group region of the membrane. Larger rings of known structure, from c9-c15 in eubacteria and chloroplasts, conserve either a lysine or an arginine residue in the equivalent position. In computer simulations of hydrated membranes containing trimethylated or unmethylated bovine c8-rings and bacterial c10- or c11-rings, the head-groups of cardiolipin molecules became associated selectively with these modified and unmodified lysine residues and with adjacent polar amino acids and with a second conserved lysine on the opposite side of the membrane, whereas phosphatidyl lipids were attracted little to these sites. However, the residence times of cardiolipin molecules with the ring were brief and sufficient for the rotor to turn only a fraction of a degree in the active enzyme. With the demethylated c8-ring and with c10- and c11-rings, the density of bound cardiolipin molecules at this site increased, but residence times were not changed greatly. These highly specific but brief interactions with the rotating c-ring are consistent with functional roles for cardiolipin in stabilizing and lubricating the rotor, and, by interacting with the enzyme at the inlet and exit of the transmembrane proton channel, in participation in proton translocation through the membrane domain of the enzyme.
Collapse
|
193
|
Gerle C. On the structural possibility of pore-forming mitochondrial FoF1 ATP synthase. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1857:1191-1196. [PMID: 26968896 DOI: 10.1016/j.bbabio.2016.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 12/31/2022]
Abstract
The mitochondrial permeability transition is an inner mitochondrial membrane event involving the opening of the permeability transition pore concomitant with a sudden efflux of matrix solutes and breakdown of membrane potential. The mitochondrial F(o)F(1) ATP synthase has been proposed as the molecular identity of the permeability transition pore. The likeliness of potential pore-forming sites in the mitochondrial F(o)F(1) ATP synthase is discussed and a new model, the death finger model, is described. In this model, movement of a p-side density that connects the lipid-plug of the c-ring with the distal membrane bending Fo domain allows reversible opening of the c-ring and structural cross-talk with OSCP and the catalytic (αβ)(3) hexamer. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
Collapse
Affiliation(s)
- Christoph Gerle
- Picobiology Institute, Department of Life Science, Graduate School of Life Science, University of Hyogo, Kamigori, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan.
| |
Collapse
|
194
|
|
195
|
Hoagland DT, Liu J, Lee RB, Lee RE. New agents for the treatment of drug-resistant Mycobacterium tuberculosis. Adv Drug Deliv Rev 2016; 102:55-72. [PMID: 27151308 PMCID: PMC4903924 DOI: 10.1016/j.addr.2016.04.026] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
Inadequate dosing and incomplete treatment regimens, coupled with the ability of the tuberculosis bacilli to cause latent infections that are tolerant of currently used drugs, have fueled the rise of multidrug-resistant tuberculosis (MDR-TB). Treatment of MDR-TB infections is a major clinical challenge that has few viable or effective solutions; therefore patients face a poor prognosis and years of treatment. This review focuses on emerging drug classes that have the potential for treating MDR-TB and highlights their particular strengths as leads including their mode of action, in vivo efficacy, and key medicinal chemistry properties. Examples include the newly approved drugs bedaquiline and delaminid, and other agents in clinical and late preclinical development pipeline for the treatment of MDR-TB. Herein, we discuss the challenges to developing drugs to treat tuberculosis and how the field has adapted to these difficulties, with an emphasis on drug discovery approaches that might produce more effective agents and treatment regimens.
Collapse
Affiliation(s)
- Daniel T Hoagland
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Pharmaceutical Sciences Graduate Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Robin B Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
196
|
Heikal A, Hards K, Cheung CY, Menorca A, Timmer MSM, Stocker BL, Cook GM. Activation of type II NADH dehydrogenase by quinolinequinones mediates antitubercular cell death. J Antimicrob Chemother 2016; 71:2840-7. [DOI: 10.1093/jac/dkw244] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/19/2016] [Indexed: 12/31/2022] Open
|
197
|
Hotra A, Suter M, Biuković G, Ragunathan P, Kundu S, Dick T, Grüber G. Deletion of a unique loop in the mycobacterial F-ATP synthase γ subunit sheds light on its inhibitory role in ATP hydrolysis-driven H(+) pumping. FEBS J 2016; 283:1947-61. [PMID: 26996828 DOI: 10.1111/febs.13715] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/29/2016] [Accepted: 03/15/2016] [Indexed: 11/28/2022]
Abstract
The F1 FO -ATP synthase is one of the enzymes that is essential to meet the energy requirement of both the proliferating aerobic and hypoxic dormant stages of the life cycle of mycobacteria. Most F-ATP synthases consume ATP in the α3 :β3 headpiece to drive the γ subunit, which couples ATP cleavage with proton pumping in the c ring of FO via the bottom of the γ subunit. ATPase-driven H(+) pumping is latent in mycobacteria. The presence of a unique 14 amino acid residue loop of the mycobacterial γ subunit has been described and aligned in close vicinity to the c-ring loop Priya R et al. (2013) J Bioenerg Biomembr 45, 121-129 Here, we used inverted membrane vesicles (IMVs) of fast-growing Mycobacterium smegmatis and a variety of covalent and non-covalent inhibitors to characterize the ATP hydrolysis activity of the F-ATP synthase inside IMVs. These vesicles formed a platform to investigate the function of the unique mycobaterial γ loop by deleting the respective loop-encoding sequence (γ166-179 ) in the genome of M. smegmatis. ATP hydrolysis-driven H(+) pumping was observed in IMVs containing the Δγ166-179 mutant protein but not for IMVs containing the wild-type F-ATP synthase. In addition, when compared to the wild-type enzyme, IMVs containing the Δγ166-179 mutant protein showed increased ATP cleavage and lower levels of ATP synthesis, demonstrating that the loop affects ATPase activity, ATPase-driven H(+) pumping and ATP synthesis. These results further indicate that the loop may affect coupling of ATP hydrolysis and synthesis in a different mode.
Collapse
Affiliation(s)
- Adam Hotra
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, Singapore.,Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore
| | - Manuel Suter
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Goran Biuković
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Priya Ragunathan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Subhashri Kundu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Thomas Dick
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gerhard Grüber
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
198
|
On the ATP binding site of the ε subunit from bacterial F-type ATP synthases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:332-40. [PMID: 26780667 DOI: 10.1016/j.bbabio.2016.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/11/2015] [Accepted: 01/14/2016] [Indexed: 11/20/2022]
Abstract
F-type ATP synthases are reversible machinery that not only synthesize adenosine triphosphate (ATP) using an electrochemical gradient across the membrane, but also can hydrolyze ATP to pump ions under certain conditions. To prevent wasteful ATP hydrolysis, subunit ε in bacterial ATP synthases changes its conformation from the non-inhibitory down- to the inhibitory up-state at a low cellular ATP concentration. Recently, a crystal structure of the ε subunit in complex with ATP was solved in a non-biologically relevant dimeric form. Here, to derive the functional ATP binding site motif, we carried out molecular dynamics simulations and free energy calculations. Our results suggest that the ATP binding site markedly differs from the experimental resolved one; we observe a reorientation of several residues, which bind to ATP in the crystal structure. In addition we find that an Mg(2+) ion is coordinated by ATP, replacing interactions of the second chain in the crystal structure. Thus we demonstrate more generally the influence of crystallization effects on ligand binding sites and their respective binding modes. Furthermore, we propose a role for two highly conserved residues to control the ATP binding/unbinding event, which have not been considered before. Additionally our results provide the basis for the rational development of new biosensors based on subunit ε, as shown previously for novel sensors measuring the ATP concentration in cells.
Collapse
|
199
|
Rombouts JA, Veenboer RP, Villellas C, Lu P, Ehlers AW, Andries K, Koul A, Lill H, Ruijter E, Orru RVA, Lammertsma K, Bald D, Slootweg JC. Synthesis, characterization and biological activity of fluorescently labeled bedaquiline analogues. RSC Adv 2016. [DOI: 10.1039/c6ra22693k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Labeling the tuberculosis drug bedaquiline with a fluorescent alkynyl-BODIPY moiety does not disrupt its antibacterial properties.
Collapse
|
200
|
Qiao CJ, Wang XK, Xie F, Zhong W, Li S. Asymmetric Synthesis and Absolute Configuration Assignment of a New Type of Bedaquiline Analogue. Molecules 2015; 20:22272-85. [PMID: 26690407 PMCID: PMC6331863 DOI: 10.3390/molecules201219846] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 12/02/2022] Open
Abstract
Bedaquiline is the first FDA-approved new chemical entity to fight multidrug-resistant tuberculosis in the last forty years. Our group replaced the quinoline ring with a naphthalene ring, leading to a new type of triarylbutanol skeleton. An asymmetric synthetic route was established for our bedaquiline analogues, and the goal of assigning their absolute configurations was achieved by comparison of experimental and calculated electronic circular dichroism spectra, and was confirmed by the combined use of circular dichroism and NMR spectroscopy.
Collapse
Affiliation(s)
- Chang-Jiang Qiao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China.
| | - Xiao-Kui Wang
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China.
| | - Fei Xie
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China.
| | - Wu Zhong
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China.
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
- Laboratory of Computer-Aided Drug Design & Discovery, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China.
| |
Collapse
|