51
|
Plasticity of the Mycobacterium tuberculosis respiratory chain and its impact on tuberculosis drug development. Nat Commun 2019; 10:4970. [PMID: 31672993 PMCID: PMC6823465 DOI: 10.1038/s41467-019-12956-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 10/09/2019] [Indexed: 12/30/2022] Open
Abstract
The viability of Mycobacterium tuberculosis (Mtb) depends on energy generated by its respiratory chain. Cytochrome bc1-aa3 oxidase and type-2 NADH dehydrogenase (NDH-2) are respiratory chain components predicted to be essential, and are currently targeted for drug development. Here we demonstrate that an Mtb cytochrome bc1-aa3 oxidase deletion mutant is viable and only partially attenuated in mice. Moreover, treatment of Mtb-infected marmosets with a cytochrome bc1-aa3 oxidase inhibitor controls disease progression and reduces lesion-associated inflammation, but most lesions become cavitary. Deletion of both NDH-2 encoding genes (Δndh-2 mutant) reveals that the essentiality of NDH-2 as shown in standard growth media is due to the presence of fatty acids. The Δndh-2 mutant is only mildly attenuated in mice and not differently susceptible to clofazimine, a drug in clinical use proposed to engage NDH-2. These results demonstrate the intrinsic plasticity of Mtb's respiratory chain, and highlight the challenges associated with targeting the pathogen's respiratory enzymes for tuberculosis drug development.
Collapse
|
52
|
Abstract
The global tuberculosis (TB) epidemic has been exacerbated by the rise in drug-resistant TB cases worldwide. To tackle this crisis, it is necessary to identify new vulnerable drug targets in Mycobacterium tuberculosis, the causative agent of TB, and develop compounds that can inhibit the bacterium through novel mechanisms of action. The QcrB subunit of the electron transport chain enzyme cytochrome bc1 has recently been validated to be a potential drug target. In the current work, we report the discovery of a new class of QcrB inhibitors, 4-amino-thieno[2,3-d]pyrimidines, that potently inhibit M. tuberculosis growth in vitro. These compounds are chemically distinct from previously reported QcrB inhibitors, and therefore, 4-amino-thieno[2,3-d]pyrimidines represent a new scaffold that can be exploited to inhibit this drug target. Antibiotic resistance is a global crisis that threatens our ability to treat bacterial infections, such as tuberculosis, caused by Mycobacterium tuberculosis. Of the 10 million cases of tuberculosis in 2017, approximately 19% of new cases and 43% of previously treated cases were caused by strains of M. tuberculosis resistant to at least one frontline antibiotic. There is a clear need for new therapies that target these genetically resistant strains. Here, we report the discovery of a new series of antimycobacterial compounds, 4-amino-thieno[2,3-d]pyrimidines, that potently inhibit the growth of M. tuberculosis. To elucidate the mechanism by which these compounds inhibit M. tuberculosis, we selected for mutants resistant to a representative 4-amino-thieno[2,3-d]pyrimidine and sequenced these strains to identify the mutations that confer resistance. We isolated a total of 12 resistant mutants, each of which harbored a nonsynonymous mutation in the gene qcrB, which encodes a subunit of the electron transport chain (ETC) enzyme cytochrome bc1 oxidoreductase, leading us to hypothesize that 4-amino-thieno[2,3-d]pyrimidines target this enzyme complex. We found that addition of 4-amino-thieno[2,3-d]pyrimidines to M. tuberculosis cultures resulted in a decrease in ATP levels, supporting our model that these compounds inhibit the M. tuberculosis ETC. Furthermore, 4-amino-thieno[2,3-d]pyrimidines had enhanced activity against a mutant of M. tuberculosis deficient in cytochrome bd oxidase, which is a hallmark of cytochrome bc1 inhibitors. Therefore, 4-amino-thieno[2,3-d]pyrimidines represent a novel series of QcrB inhibitors that build on the growing number of chemical scaffolds that are able to inhibit the mycobacterial cytochrome bc1 complex. IMPORTANCE The global tuberculosis (TB) epidemic has been exacerbated by the rise in drug-resistant TB cases worldwide. To tackle this crisis, it is necessary to identify new vulnerable drug targets in Mycobacterium tuberculosis, the causative agent of TB, and develop compounds that can inhibit the bacterium through novel mechanisms of action. The QcrB subunit of the electron transport chain enzyme cytochrome bc1 has recently been validated to be a potential drug target. In the current work, we report the discovery of a new class of QcrB inhibitors, 4-amino-thieno[2,3-d]pyrimidines, that potently inhibit M. tuberculosis growth in vitro. These compounds are chemically distinct from previously reported QcrB inhibitors, and therefore, 4-amino-thieno[2,3-d]pyrimidines represent a new scaffold that can be exploited to inhibit this drug target.
Collapse
|
53
|
Campaniço A, Carrasco MP, Njoroge M, Seldon R, Chibale K, Perdigão J, Portugal I, Warner DF, Moreira R, Lopes F. Azaaurones as Potent Antimycobacterial Agents Active against MDR- and XDR-TB. ChemMedChem 2019; 14:1537-1546. [PMID: 31294529 DOI: 10.1002/cmdc.201900289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/19/2019] [Indexed: 12/31/2022]
Abstract
Herein we report the screening of a small library of aurones and their isosteric counterparts, azaaurones and N-acetylazaaurones, against Mycobacterium tuberculosis. Aurones were found to be inactive at 20 μm, whereas azaaurones and N-acetylazaaurones emerged as the most potent compounds, with nine derivatives displaying MIC99 values ranging from 0.4 to 2.0 μm. In addition, several N-acetylazaaurones were found to be active against multidrug-resistant (MDR) and extensively drug-resistant (XDR) clinical M. tuberculosis isolates. The antimycobacterial mechanism of action of these compounds remains to be determined; however, a preliminary mechanistic study confirmed that they do not inhibit the mycobacterial cytochrome bc1 complex. Additionally, microsomal metabolic stability and metabolite identification studies revealed that N-acetylazaaurones are deacetylated to their azaaurone counterparts. Overall, these results demonstrate that azaaurones and their N-acetyl counterparts represent a new entry in the toolbox of chemotypes capable of inhibiting M. tuberculosis growth.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Marta P Carrasco
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Mathew Njoroge
- Division of Clinical Pharmacology, Department of Medicine, Drug Discovery and Development Centre (H3D), University of Cape Town, Observatory, 7925, South Africa
| | - Ronnett Seldon
- Department of Chemistry, South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa.,Department of Chemistry, South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | - João Perdigão
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Isabel Portugal
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Digby F Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa.,Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch, 7701, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
54
|
Kalia NP, Shi Lee B, Ab Rahman NB, Moraski GC, Miller MJ, Pethe K. Carbon metabolism modulates the efficacy of drugs targeting the cytochrome bc 1:aa 3 in Mycobacterium tuberculosis. Sci Rep 2019; 9:8608. [PMID: 31197236 PMCID: PMC6565617 DOI: 10.1038/s41598-019-44887-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/23/2019] [Indexed: 11/30/2022] Open
Abstract
The influence of carbon metabolism on oxidative phosphorylation is poorly understood in mycobacteria. M. tuberculosis expresses two respiratory terminal oxidases, the cytochrome bc1:aa3 and the cytochrome bd oxidase, which are jointly required for oxidative phosphorylation and mycobacterial viability. The essentiality of the cytochrome bc1:aa3 for optimum growth is illustrated by its vulnerability to chemical inhibition by the clinical drug candidate Q203 and several other chemical series. The cytochrome bd oxidase is not strictly essential for growth but is required to maintain bioenergetics when the function of the cytochrome bc1:aa3 is compromised. In this study, we observed that the potency of drugs targeting the cytochrome bc1:aa3 is influenced by carbon metabolism. The efficacy of Q203 and related derivatives was alleviated by glycerol supplementation. The negative effect of glycerol supplementation on Q203 potency correlated with an upregulation of the cytochrome bd oxidase-encoding cydABDC operon. Upon deletion of cydAB, the detrimental effect of glycerol on the potency of Q203 was abrogated. The same phenomenon was also observed in recent clinical isolates, but to a lesser extent compared to the laboratory-adapted strain H37Rv. This study reinforces the importance of optimizing in vitro culture conditions for drug evaluation in mycobacteria, a factor which appeared to be particularly essential for drugs targeting the cytochrome bc1:aa3 terminal oxidase.
Collapse
Affiliation(s)
- Nitin P Kalia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Bei Shi Lee
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Nurlilah B Ab Rahman
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Garrett C Moraski
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Marvin J Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
55
|
Novel MenA Inhibitors Are Bactericidal against Mycobacterium tuberculosis and Synergize with Electron Transport Chain Inhibitors. Antimicrob Agents Chemother 2019; 63:AAC.02661-18. [PMID: 30962346 PMCID: PMC6535543 DOI: 10.1128/aac.02661-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/28/2019] [Indexed: 01/13/2023] Open
Abstract
Mycobacterium tuberculosis is the leading cause of morbidity and death resulting from infectious disease worldwide. The incredible disease burden, combined with the long course of drug treatment and an increasing incidence of antimicrobial resistance among M. tuberculosis isolates, necessitates novel drugs and drug targets for treatment of this deadly pathogen. Mycobacterium tuberculosis is the leading cause of morbidity and death resulting from infectious disease worldwide. The incredible disease burden, combined with the long course of drug treatment and an increasing incidence of antimicrobial resistance among M. tuberculosis isolates, necessitates novel drugs and drug targets for treatment of this deadly pathogen. Recent work has produced several promising clinical candidates targeting components of the electron transport chain (ETC) of M. tuberculosis, highlighting this pathway’s potential as a drug target. Menaquinone is an essential component of the M. tuberculosis ETC, as it functions to shuttle electrons through the ETC to produce the electrochemical gradient required for ATP production for the cell. We show that inhibitors of MenA, a component of the menaquinone biosynthetic pathway, are highly active against M. tuberculosis. MenA inhibitors are bactericidal against M. tuberculosis under both replicating and nonreplicating conditions, with 10-fold higher bactericidal activity against nutrient-starved bacteria than against replicating cultures. MenA inhibitors have enhanced activity in combination with bedaquiline, clofazimine, and inhibitors of QcrB, a component of the cytochrome bc1 oxidase. Together, these data support MenA as a viable target for drug treatment against M. tuberculosis. MenA inhibitors not only kill M. tuberculosis in a variety of physiological states but also show enhanced activity in combination with ETC inhibitors in various stages of clinical trial testing.
Collapse
|
56
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
57
|
Scherr N, Bieri R, Thomas SS, Chauffour A, Kalia NP, Schneide P, Ruf MT, Lamelas A, Manimekalai MSS, Grüber G, Ishii N, Suzuki K, Tanner M, Moraski GC, Miller MJ, Witschel M, Jarlier V, Pluschke G, Pethe K. Targeting the Mycobacterium ulcerans cytochrome bc 1:aa 3 for the treatment of Buruli ulcer. Nat Commun 2018; 9:5370. [PMID: 30560872 PMCID: PMC6299076 DOI: 10.1038/s41467-018-07804-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/26/2018] [Indexed: 11/21/2022] Open
Abstract
Mycobacterium ulcerans is the causative agent of Buruli ulcer, a neglected tropical skin disease that is most commonly found in children from West and Central Africa. Despite the severity of the infection, therapeutic options are limited to antibiotics with severe side effects. Here, we show that M. ulcerans is susceptible to the anti-tubercular drug Q203 and related compounds targeting the respiratory cytochrome bc1:aa3. While the cytochrome bc1:aa3 is the primary terminal oxidase in Mycobacterium tuberculosis, the presence of an alternate bd-type terminal oxidase limits the bactericidal and sterilizing potency of Q203 against this bacterium. M. ulcerans strains found in Buruli ulcer patients from Africa and Australia lost all alternate terminal electron acceptors and rely exclusively on the cytochrome bc1:aa3 to respire. As a result, Q203 is bactericidal at low dose against M. ulcerans replicating in vitro and in mice, making the drug a promising candidate for Buruli ulcer treatment. Mycobacterium ulcerans is the causative agent of Buruli ulcer (BU). Existing anti-tubercular drugs have been used to treat the condition with varying success. Here, the authors show that a clinical-stage drug candidate for tuberculosis, Q203, is effective at killing M. ulcerans and is a promising therapeutic candidate for BU.
Collapse
Affiliation(s)
- Nicole Scherr
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland.,University of Basel, Basel, 4001, Switzerland
| | - Raphael Bieri
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland.,University of Basel, Basel, 4001, Switzerland
| | - Sangeeta S Thomas
- Lee Kong Chian School of Medicine, Nanyang Technological University, Experimental Medicine Building, Singapore, 636921, Singapore
| | - Aurélie Chauffour
- CR7, INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses, CIMI, Team E13 (Bactériologie), Sorbonne Universités, UPMC Université Paris 06, Paris, 75005, France
| | - Nitin Pal Kalia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Experimental Medicine Building, Singapore, 636921, Singapore
| | | | - Marie-Thérèse Ruf
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland.,University of Basel, Basel, 4001, Switzerland
| | - Araceli Lamelas
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland.,University of Basel, Basel, 4001, Switzerland.,Red de Estudios Moleculares, AvanzadosInstituto de Ecología A. C., Xalapa, 91000, Veracruz, Mexico
| | - Malathy S S Manimekalai
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Gerhard Grüber
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Norihisa Ishii
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, 189-0002, Japan
| | - Koichi Suzuki
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, 189-0002, Japan.,Department of Clinical Laboratory Science, Faculty of Medical Technology, Teikyo University, Tokyo, 173-8605, Japan
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland.,University of Basel, Basel, 4001, Switzerland
| | - Garrett C Moraski
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59715, USA
| | - Marvin J Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | | | - Vincent Jarlier
- CR7, INSERM, U1135, Centre d'Immunologie et des Maladies Infectieuses, CIMI, Team E13 (Bactériologie), Sorbonne Universités, UPMC Université Paris 06, Paris, 75005, France.,CNR-MyRMA, Bactériologie Hygiène, Hôpitaux Universitaires Pitie Salpêtrière-Charles Foix, Paris, 75013, France
| | - Gerd Pluschke
- Swiss Tropical and Public Health Institute, Basel, 4051, Switzerland. .,University of Basel, Basel, 4001, Switzerland.
| | - Kevin Pethe
- Lee Kong Chian School of Medicine, Nanyang Technological University, Experimental Medicine Building, Singapore, 636921, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
58
|
Xu Z, Zhou A, Wu J, Zhou A, Li J, Zhang S, Wu W, Karakousis PC, Yao YF. Transcriptional Approach for Decoding the Mechanism of rpoC Compensatory Mutations for the Fitness Cost in Rifampicin-Resistant Mycobacterium tuberculosis. Front Microbiol 2018; 9:2895. [PMID: 30555440 PMCID: PMC6283890 DOI: 10.3389/fmicb.2018.02895] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 11/12/2018] [Indexed: 12/31/2022] Open
Abstract
Multidrug-resistant tuberculosis (TB), defined as TB resistant to the two first-line drugs, isoniazid and rifampin, is a serious challenge to global TB eradication efforts. Although mutations in rpoA or rpoC have been proposed to compensate for this fitness cost due to rpoB mutation in rifampicin-resistant Mycobacterium tuberculosis mutants, whether the compensatory effect exists and the underlying mechanisms of compensation remain unclear. Here, we used RNA sequencing to investigate the global transcriptional profiles of 6 rifampin-resistant clinical isolates with either single mutation in rpoB or dual mutations in rpoB/rpoC, as well as 3 rifampin-susceptible clinical isolates, trying to prove the potential compensatory effect of rpoC by transcriptomic alteration. In rifampin-free conditions, rpoC mutation was associated with M. tuberculosis upregulation of ribosomal protein-coding genes, dysregulation of growth-related essential genes and balancing the expression of arginine and glutamate synthesis-associated genes. Upon rifampin exposure of M. tuberculosis isolates, rpoC mutations were associated with the upregulation of the oxidative phosphorylation machinery, which was inhibited in the rpoB single mutants, as well as stabilization of the expression of rifampin-regulated essential genes and balancing the expression of genes involved in metabolism of sulfur-containing amino acids. Taken together, our data suggest that rpoC mutation may compensate for the fitness defect of rifampicin-resistant M. tuberculosis by altering gene expression in response to rifampin exposure.
Collapse
Affiliation(s)
- Zhihong Xu
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiping Zhou
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiawei Wu
- Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiwu Zhou
- Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Shulin Zhang
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Petros C Karakousis
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
59
|
Oh S, Park Y, Engelhart CA, Wallach JB, Schnappinger D, Arora K, Manikkam M, Gac B, Wang H, Murgolo N, Olsen DB, Goodwin M, Sutphin M, Weiner DM, Via LE, Boshoff HIM, Barry CE. Discovery and Structure-Activity-Relationship Study of N-Alkyl-5-hydroxypyrimidinone Carboxamides as Novel Antitubercular Agents Targeting Decaprenylphosphoryl-β-d-ribose 2'-Oxidase. J Med Chem 2018; 61:9952-9965. [PMID: 30350998 PMCID: PMC6257622 DOI: 10.1021/acs.jmedchem.8b00883] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Magnesium plays an important role
in infection with Mycobacterium
tuberculosis (Mtb) as a signal of the extracellular
environment, as a cofactor for many enzymes, and as a structural element
in important macromolecules. Raltegravir, an antiretroviral drug that
inhibits HIV-1 integrase is known to derive its potency from selective
sequestration of active-site magnesium ions in addition to binding
to a hydrophobic pocket. In order to determine if essential Mtb-related phosphoryl transfers could be disrupted in a
similar manner, a directed screen of known molecules with integrase
inhibitor-like pharmacophores (N-alkyl-5-hydroxypyrimidinone
carboxamides) was performed. Initial hits afforded compounds with
low-micromolar potency against Mtb, acceptable cytotoxicity
and PK characteristics, and robust SAR. Elucidation of the target
of these compounds revealed that they lacked magnesium dependence
and instead disappointingly inhibited a known promiscuous target in Mtb, decaprenylphosphoryl-β-d-ribose 2′-oxidase
(DprE1, Rv3790).
Collapse
Affiliation(s)
- Sangmi Oh
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Yumi Park
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Curtis A Engelhart
- Department of Microbiology and Immunology , Weill Cornell Medical College , New York , New York 10021 , United States
| | - Joshua B Wallach
- Department of Microbiology and Immunology , Weill Cornell Medical College , New York , New York 10021 , United States
| | - Dirk Schnappinger
- Department of Microbiology and Immunology , Weill Cornell Medical College , New York , New York 10021 , United States
| | - Kriti Arora
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Michelle Manikkam
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Brian Gac
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Hongwu Wang
- Discovery Research , Merck & Company, Inc. , 770 Sumneytown Pike , West Point , Pennsylvania 19486 , United States
| | - Nicholas Murgolo
- Discovery Research , Merck & Company, Inc. , 770 Sumneytown Pike , West Point , Pennsylvania 19486 , United States
| | - David B Olsen
- Discovery Research , Merck & Company, Inc. , 770 Sumneytown Pike , West Point , Pennsylvania 19486 , United States
| | - Michael Goodwin
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Michelle Sutphin
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Danielle M Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States.,Institute for Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town 7935 , South Africa
| | - Helena I M Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Bethesda , Maryland 20892 , United States.,Institute for Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town 7935 , South Africa
| |
Collapse
|
60
|
No Evidence for Acquired Mutations Associated with Cytochrome bc 1 Inhibitor Resistance in 13,559 Clinical Mycobacterium tuberculosis Complex Isolates. Antimicrob Agents Chemother 2018; 63:AAC.01317-18. [PMID: 30323041 PMCID: PMC6325196 DOI: 10.1128/aac.01317-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
61
|
Foo CS, Lupien A, Kienle M, Vocat A, Benjak A, Sommer R, Lamprecht DA, Steyn AJC, Pethe K, Piton J, Altmann KH, Cole ST. Arylvinylpiperazine Amides, a New Class of Potent Inhibitors Targeting QcrB of Mycobacterium tuberculosis. mBio 2018; 9:e01276-18. [PMID: 30301850 PMCID: PMC6178619 DOI: 10.1128/mbio.01276-18] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 11/20/2022] Open
Abstract
New drugs are needed to control the current tuberculosis (TB) pandemic caused by infection with Mycobacterium tuberculosis We report here on our work with AX-35, an arylvinylpiperazine amide, and four related analogs, which are potent antitubercular agents in vitro All five compounds showed good activity against M. tuberculosisin vitro and in infected THP-1 macrophages, while displaying only mild cytotoxicity. Isolation and characterization of M. tuberculosis-resistant mutants to the arylvinylpiperazine amide derivative AX-35 revealed mutations in the qcrB gene encoding a subunit of cytochrome bc1 oxidase, one of two terminal oxidases of the electron transport chain. Cross-resistance studies, allelic exchange, transcriptomic analyses, and bioenergetic flux assays provided conclusive evidence that the cytochrome bc1-aa3 is the target of AX-35, although the compound appears to interact differently with the quinol binding pocket compared to previous QcrB inhibitors. The transcriptomic and bioenergetic profiles of M. tuberculosis treated with AX-35 were similar to those generated by other cytochrome bc1 oxidase inhibitors, including the compensatory role of the alternate terminal oxidase cytochrome bd in respiratory adaptation. In the absence of cytochrome bd oxidase, AX-35 was bactericidal against M. tuberculosis Finally, AX-35 and its analogs were active in an acute mouse model of TB infection, with two analogs displaying improved activity over the parent compound. Our findings will guide future lead optimization to produce a drug candidate for the treatment of TB and other mycobacterial diseases, including Buruli ulcer and leprosy.IMPORTANCE New drugs against Mycobacterium tuberculosis are urgently needed to deal with the current global TB pandemic. We report here on the discovery of a series of arylvinylpiperazine amides (AX-35 to AX-39) that represent a promising new family of compounds with potent in vitro and in vivo activities against M. tuberculosis AX compounds target the QcrB subunit of the cytochrome bc1 terminal oxidase with a different mode of interaction compared to those of known QcrB inhibitors. This study provides the first multifaceted validation of QcrB inhibition by recombineering-mediated allelic exchange, gene expression profiling, and bioenergetic flux studies. It also provides further evidence for the compensatory role of cytochrome bd oxidase upon QcrB inhibition. In the absence of cytochrome bd oxidase, AX compounds are bactericidal, an encouraging property for future antimycobacterial drug development.
Collapse
Affiliation(s)
- Caroline S Foo
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Andréanne Lupien
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maryline Kienle
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Anthony Vocat
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Andrej Benjak
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Raphael Sommer
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Adrie J C Steyn
- Africa Health Research Institute, Durban, South Africa
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jérémie Piton
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Stewart T Cole
- Global Health Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
62
|
Beteck RM, Seldon R, Coertzen D, van der Watt ME, Reader J, Mackenzie JS, Lamprecht DA, Abraham M, Eribez K, Müller J, Rui F, Zhu G, de Grano RV, Williams ID, Smit FJ, Steyn AJC, Winzeler EA, Hemphill A, Birkholtz LM, Warner DF, N’Da DD, Haynes RK. Accessible and distinct decoquinate derivatives active against Mycobacterium tuberculosis and apicomplexan parasites. Commun Chem 2018. [DOI: 10.1038/s42004-018-0062-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
63
|
J Libardo MD, Boshoff HI, Barry CE. The present state of the tuberculosis drug development pipeline. Curr Opin Pharmacol 2018; 42:81-94. [PMID: 30144650 DOI: 10.1016/j.coph.2018.08.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 10/28/2022]
Abstract
Tuberculosis now ranks as the leading cause of death in the world due to a single infectious agent. Current standard of care treatment can achieve very high cure rates for drug-sensitive disease but requires a 6-month duration of chemotherapy. Drug-resistant disease requires significantly longer treatment durations with drugs associated with a higher risk of adverse events. Thus, there is a pressing need for a drug regimen that is safer, shorter in duration and superior to current front-line chemotherapy in terms of efficacy. The TB drug pipeline contains several candidates that address one or more of the required attributes of chemotherapeutic regimens that may redefine the standard of care of this disease. Several new drugs have been reported and novel targets have been identified allowing regimens containing new compounds to trickle into clinical studies. Furthermore, a recent paradigm-shift in understanding the pharmacokinetics of anti-tubercular drugs is revolutionizing the way we select compounds for clinical progression.
Collapse
Affiliation(s)
- M Daben J Libardo
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Helena Im Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
64
|
Cleghorn LAT, Ray PC, Odingo J, Kumar A, Wescott H, Korkegian A, Masquelin T, Lopez Moure A, Wilson C, Davis S, Huggett M, Turner P, Smith A, Epemolu O, Zuccotto F, Riley J, Scullion P, Shishikura Y, Ferguson L, Rullas J, Guijarro L, Read KD, Green SR, Hipskind P, Parish T, Wyatt PG. Identification of Morpholino Thiophenes as Novel Mycobacterium tuberculosis Inhibitors, Targeting QcrB. J Med Chem 2018; 61:6592-6608. [PMID: 29944372 PMCID: PMC6089501 DOI: 10.1021/acs.jmedchem.8b00172] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
With the emergence of multidrug-resistant strains of Mycobacterium tuberculosis there is a pressing need for new oral drugs with novel mechanisms of action. Herein, we describe the identification of a novel morpholino-thiophenes (MOT) series following phenotypic screening of the Eli Lilly corporate library against M. tuberculosis strain H37Rv. The design, synthesis, and structure-activity relationships of a range of analogues around the confirmed actives are described. Optimized leads with potent whole cell activity against H37Rv, no cytotoxicity flags, and in vivo efficacy in an acute murine model of infection are described. Mode-of-action studies suggest that the novel scaffold targets QcrB, a subunit of the menaquinol cytochrome c oxidoreductase, part of the bc1-aa3-type cytochrome c oxidase complex that is responsible for driving oxygen-dependent respiration.
Collapse
Affiliation(s)
- Laura A T Cleghorn
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Peter C Ray
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Joshua Odingo
- TB Discovery Research , Infectious Disease Research Institute (IDRI) , 1616 Eastlake Avenue East, Suite 400 , Seattle , Washington 98102 , United States
| | - Anuradha Kumar
- TB Discovery Research , Infectious Disease Research Institute (IDRI) , 1616 Eastlake Avenue East, Suite 400 , Seattle , Washington 98102 , United States
| | - Heather Wescott
- TB Discovery Research , Infectious Disease Research Institute (IDRI) , 1616 Eastlake Avenue East, Suite 400 , Seattle , Washington 98102 , United States
| | - Aaron Korkegian
- TB Discovery Research , Infectious Disease Research Institute (IDRI) , 1616 Eastlake Avenue East, Suite 400 , Seattle , Washington 98102 , United States
| | - Thierry Masquelin
- Eli Lilly and Company, Discovery Chemistry Research, Lilly Corporate Centre , MC/87/02/203, G17, Indianapolis , Indiana 46285 , United States
| | - Abraham Lopez Moure
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Caroline Wilson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Susan Davis
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Margaret Huggett
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Penelope Turner
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Alasdair Smith
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Ola Epemolu
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Fabio Zuccotto
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Paul Scullion
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Yoko Shishikura
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Liam Ferguson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Joaquin Rullas
- Diseases of the Developing World , GlaxoSmithKline , Calle Severo Ochoa 2 , 28760 Tres Cantos , Madrid Spain
| | - Laura Guijarro
- Diseases of the Developing World , GlaxoSmithKline , Calle Severo Ochoa 2 , 28760 Tres Cantos , Madrid Spain
| | - Kevin D Read
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Simon R Green
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| | - Phil Hipskind
- Eli Lilly and Company, Discovery Chemistry Research, Lilly Corporate Centre , MC/87/02/203, G17, Indianapolis , Indiana 46285 , United States
| | - Tanya Parish
- TB Discovery Research , Infectious Disease Research Institute (IDRI) , 1616 Eastlake Avenue East, Suite 400 , Seattle , Washington 98102 , United States
| | - Paul G Wyatt
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences , University of Dundee , Dundee DD1 5EH , United Kingdom
| |
Collapse
|
65
|
Lee BS, Pethe K. Therapeutic potential of promiscuous targets in Mycobacterium tuberculosis. Curr Opin Pharmacol 2018; 42:22-26. [PMID: 30015177 DOI: 10.1016/j.coph.2018.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/31/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022]
Abstract
In the field of tuberculosis drug development, the term 'promiscuous' was coined to collectively describe targets that repeatedly show up in whole-cell screenings. With the current climate leaning towards the exclusion of these targets in future drug screens, this review discusses and clarifies misconceptions surrounding this classification, the prospects of developing compounds targeting promiscuous targets, and their potential impact on tuberculosis drug development. The dominance of these targets in cell-based screens reflect not only bias introduced by experimental setup, but also some of the pathogen's greatest vulnerabilities. Coupled with favourable predictions of their in vivo efficacies and synergism with other TB drugs, these targets open opportunities to be explored for the development of rational drug combination for tuberculosis.
Collapse
Affiliation(s)
- Bei Shi Lee
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kevin Pethe
- School of Biological Sciences, Nanyang Technological University, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Experimental Medicine Building, Singapore.
| |
Collapse
|
66
|
Roles of Alanine Dehydrogenase and Induction of Its Gene in Mycobacterium smegmatis under Respiration-Inhibitory Conditions. J Bacteriol 2018; 200:JB.00152-18. [PMID: 29712875 DOI: 10.1128/jb.00152-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/25/2018] [Indexed: 01/08/2023] Open
Abstract
Here we demonstrated that the inhibition of electron flux through the respiratory electron transport chain (ETC) by either the disruption of the gene for the major terminal oxidase (aa3 cytochrome c oxidase) or treatment with KCN resulted in the induction of ald encoding alanine dehydrogenase in Mycobacterium smegmatis A decrease in functionality of the ETC shifts the redox state of the NADH/NAD+ pool toward a more reduced state, which in turn leads to an increase in cellular levels of alanine by Ald catalyzing the conversion of pyruvate to alanine with the concomitant oxidation of NADH to NAD+ The induction of ald expression under respiration-inhibitory conditions in M. smegmatis is mediated by the alanine-responsive AldR transcriptional regulator. The growth defect of M. smegmatis by respiration inhibition was exacerbated by inactivation of the ald gene, suggesting that Ald is beneficial to M. smegmatis in its adaptation and survival under respiration-inhibitory conditions by maintaining NADH/NAD+ homeostasis. The low susceptibility of M. smegmatis to bcc1 complex inhibitors appears to be, at least in part, attributable to the high expression level of the bd quinol oxidase in M. smegmatis when the bcc1-aa3 branch of the ETC is inactivated.IMPORTANCE We demonstrated that the functionality of the respiratory electron transport chain is inversely related to the expression level of the ald gene encoding alanine dehydrogenase in Mycobacterium smegmatis Furthermore, the importance of Ald in NADH/NAD+ homeostasis during the adaptation of M. smegmatis to severe respiration-inhibitory conditions was demonstrated in this study. On the basis of these results, we propose that combinatory regimens including both an Ald-specific inhibitor and respiration-inhibitory antitubercular drugs such as Q203 and bedaquiline are likely to enable a more efficient therapy for tuberculosis.
Collapse
|
67
|
Murugesan D, Ray PC, Bayliss T, Prosser GA, Harrison JR, Green K, Soares de Melo C, Feng TS, Street LJ, Chibale K, Warner DF, Mizrahi V, Epemolu O, Scullion P, Ellis L, Riley J, Shishikura Y, Ferguson L, Osuna-Cabello M, Read KD, Green SR, Lamprecht DA, Finin PM, Steyn AJC, Ioerger TR, Sacchettini J, Rhee KY, Arora K, Barry CE, Wyatt PG, Boshoff HIM. 2-Mercapto-Quinazolinones as Inhibitors of Type II NADH Dehydrogenase and Mycobacterium tuberculosis: Structure-Activity Relationships, Mechanism of Action and Absorption, Distribution, Metabolism, and Excretion Characterization. ACS Infect Dis 2018. [PMID: 29522317 PMCID: PMC5996347 DOI: 10.1021/acsinfecdis.7b00275] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Mycobacterium tuberculosis (MTb) possesses
two nonproton pumping type II NADH dehydrogenase (NDH-2)
enzymes which are predicted to be jointly essential for respiratory
metabolism. Furthermore, the structure of a closely related bacterial
NDH-2 has been reported recently, allowing for the structure-based
design of small-molecule inhibitors. Herein, we disclose MTb whole-cell structure–activity relationships (SARs) for a series of 2-mercapto-quinazolinones which target the ndh encoded NDH-2 with nanomolar potencies. The compounds were inactivated by glutathione-dependent adduct formation as well as quinazolinone oxidation in microsomes. Pharmacokinetic studies demonstrated modest bioavailability and compound exposures. Resistance to the compounds in MTb was conferred by promoter mutations in the alternative nonessential NDH-2 encoded by ndhA in MTb. Bioenergetic analyses revealed a decrease in oxygen consumption rates in response to inhibitor in cells in which membrane potential was uncoupled from ATP production, while inverted membrane vesicles showed mercapto-quinazolinone-dependent inhibition of ATP production when NADH was the electron donor to the respiratory chain. Enzyme kinetic studies further demonstrated noncompetitive inhibition, suggesting binding of this scaffold to an allosteric site. In summary, while the initial MTb SAR showed limited improvement in potency, these results, combined with structural information on the bacterial protein, will aid in the future discovery of new and improved NDH-2 inhibitors.
Collapse
Affiliation(s)
- Dinakaran Murugesan
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Peter C. Ray
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Tracy Bayliss
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Gareth A. Prosser
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Justin R. Harrison
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Kirsteen Green
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Candice Soares de Melo
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Tzu-Shean Feng
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Leslie J. Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Kelly Chibale
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| | - Valerie Mizrahi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| | - Ola Epemolu
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Paul Scullion
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Lucy Ellis
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Yoko Shishikura
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Liam Ferguson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Maria Osuna-Cabello
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Kevin D. Read
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Simon R. Green
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Dirk A. Lamprecht
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Peter M. Finin
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
- Department of Microbiology, University of Alabama at Birmingham, 1720 Second Avenue South, Birmingham, Alabama 35294-2170, United States
| | - Thomas R. Ioerger
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Jim Sacchettini
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Kyu Y. Rhee
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medical College, New York, New York 10065, United States
| | - Kriti Arora
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Clifton E. Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | - Paul G. Wyatt
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Helena I. M. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| |
Collapse
|
68
|
Campaniço A, Moreira R, Lopes F. Drug discovery in tuberculosis. New drug targets and antimycobacterial agents. Eur J Med Chem 2018; 150:525-545. [DOI: 10.1016/j.ejmech.2018.03.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 01/24/2023]
|
69
|
Activity of 2-(quinolin-4-yloxy)acetamides in Mycobacterium tuberculosis clinical isolates and identification of their molecular target by whole-genome sequencing. Int J Antimicrob Agents 2018; 51:378-384. [DOI: 10.1016/j.ijantimicag.2017.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 08/10/2017] [Accepted: 08/15/2017] [Indexed: 11/24/2022]
|
70
|
Lu P, Asseri AH, Kremer M, Maaskant J, Ummels R, Lill H, Bald D. The anti-mycobacterial activity of the cytochrome bcc inhibitor Q203 can be enhanced by small-molecule inhibition of cytochrome bd. Sci Rep 2018; 8:2625. [PMID: 29422632 PMCID: PMC5805707 DOI: 10.1038/s41598-018-20989-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/23/2018] [Indexed: 11/09/2022] Open
Abstract
Mycobacterial energy metabolism currently attracts strong attention as new target space for development of anti-tuberculosis drugs. The imidazopyridine Q203 targets the cytochrome bcc complex of the respiratory chain, a key component in energy metabolism. Q203 blocks growth of Mycobacterium tuberculosis at nanomolar concentrations, however, it fails to actually kill the bacteria, which may limit the clinical applicability of this candidate drug. In this report we show that inhibition of cytochrome bd, a parallel branch of the mycobacterial respiratory chain, by aurachin D invoked bactericidal activity of Q203. In biochemical assays using inverted membrane vesicles from Mycobacterium tuberculosis and Mycobacterium smegmatis we found that inhibition of respiratory chain activity by Q203 was incomplete, but could be enhanced by inactivation of cytochrome bd, either by genetic knock-out or by inhibition with aurachin D. These results indicate that simultaneously targeting the cytochrome bcc and the cytochrome bd branch of the mycobacterial respiratory chain may turn out as effective strategy for combating M. tuberculosis.
Collapse
Affiliation(s)
- Ping Lu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Amer H Asseri
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.,Biochemsitry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Martijn Kremer
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Janneke Maaskant
- Department of Medical Microbiology and Infection Control, VU university Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Roy Ummels
- Department of Medical Microbiology and Infection Control, VU university Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Holger Lill
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Dirk Bald
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
| |
Collapse
|
71
|
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]
|
72
|
Combinations of Respiratory Chain Inhibitors Have Enhanced Bactericidal Activity against Mycobacterium tuberculosis. Antimicrob Agents Chemother 2017; 62:AAC.01677-17. [PMID: 29061760 PMCID: PMC5740367 DOI: 10.1128/aac.01677-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/17/2017] [Indexed: 11/20/2022] Open
Abstract
As an obligate aerobe, Mycobacterium tuberculosis uses its electron transport chain (ETC) to produce energy via oxidative phosphorylation. This pathway has recently garnered a lot of attention and is a target for several new antimycobacterials. We tested the respiratory adaptation of M. tuberculosis to phenoxyalkylbenzimidazoles (PABs), compounds proposed to target QcrB, a component of the cytochrome bc1 complex. We show that M. tuberculosis is able to reroute its ETC to provide temporary resistance to PABs. However, combination treatment of PAB with agents targeting other components of the electron transport chain overcomes this respiratory flexibility. PAB in combination with clofazimine resulted in synergistic killing of M. tuberculosis under both replicating and nonreplicating conditions. PABs in combination with bedaquiline demonstrated antagonism at early time points, particularly under nonreplicating conditions. However, this antagonistic effect disappeared within 3 weeks, when PAB-BDQ combinations became highly bactericidal; in some cases, they were better than either drug alone. This study highlights the potential for combination treatment targeting the ETC and supports the development of PABs as part of a novel drug regimen against M. tuberculosis.
Collapse
|
73
|
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
|
74
|
Chandrasekera NS, Berube BJ, Shetye G, Chettiar S, O’Malley T, Manning A, Flint L, Awasthi D, Ioerger TR, Sacchettini J, Masquelin T, Hipskind PA, Odingo J, Parish T. Improved Phenoxyalkylbenzimidazoles with Activity against Mycobacterium tuberculosis Appear to Target QcrB. ACS Infect Dis 2017; 3:898-916. [PMID: 29035551 PMCID: PMC5727484 DOI: 10.1021/acsinfecdis.7b00112] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
The phenoxy alkyl benzimidazoles
(PABs) have good antitubercular activity. We expanded our structure–activity
relationship studies to determine the core components of PABs required
for activity. The most potent compounds had minimum inhibitory concentrations
against Mycobacterium tuberculosis in the low nanomolar
range with very little cytotoxicity against eukaryotic cells as well
as activity against intracellular bacteria. We isolated resistant
mutants against PAB compounds, which had mutations in either Rv1339,
of unknown function, or qcrB, a component of the
cytochrome bc1 oxidase of the electron
transport chain. QcrB mutant strains were resistant to all PAB compounds,
whereas Rv1339 mutant strains were only resistant to a subset, suggesting
that QcrB is the target. The discovery of the target for PAB compounds
will allow for the improved design of novel compounds to target intracellular M. tuberculosis.
Collapse
Affiliation(s)
- N. Susantha Chandrasekera
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Bryan J. Berube
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Gauri Shetye
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Somsundaram Chettiar
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Theresa O’Malley
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Alyssa Manning
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Lindsay Flint
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Divya Awasthi
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | | | | | - Thierry Masquelin
- Lilly Research Laboratories, 307 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Philip A. Hipskind
- Lilly Research Laboratories, 307 East Merrill Street, Indianapolis, Indiana 46285, United States
| | - Joshua Odingo
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| | - Tanya Parish
- TB Discovery Research, Infectious Disease Research Institute, 1616 Eastlake Avenue East, Seattle, Washington 98102, United States
| |
Collapse
|
75
|
Wilson CR, Gessner RK, Moosa A, Seldon R, Warner DF, Mizrahi V, Soares de Melo C, Simelane SB, Nchinda A, Abay E, Taylor D, Njoroge M, Brunschwig C, Lawrence N, Boshoff HIM, Barry CE, Sirgel FA, van Helden P, Harris CJ, Gordon R, Ghidelli-Disse S, Pflaumer H, Boesche M, Drewes G, Sanz O, Santos G, Rebollo-Lopez MJ, Urones B, Selenski C, Lafuente-Monasterio MJ, Axtman M, Lelièvre J, Ballell L, Mueller R, Street LJ, Ghorpade SR, Chibale K. Novel Antitubercular 6-Dialkylaminopyrimidine Carboxamides from Phenotypic Whole-Cell High Throughput Screening of a SoftFocus Library: Structure-Activity Relationship and Target Identification Studies. J Med Chem 2017; 60:10118-10134. [PMID: 29148755 PMCID: PMC5748279 DOI: 10.1021/acs.jmedchem.7b01347] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
A BioFocus
DPI SoftFocus library of ∼35 000 compounds was screened
against Mycobacterium tuberculosis (Mtb) in order
to identify novel hits with antitubercular activity. The hits were
evaluated in biology triage assays to exclude compounds suggested to function via frequently encountered promiscuous mechanisms of action including inhibition of the QcrB subunit of the cytochrome bc1 complex, disruption of cell–wall homeostasis, and DNA damage. Among the hits that passed this screening cascade, a 6-dialkylaminopyrimidine carboxamide series was prioritized for hit to lead optimization. Compounds from this series were active against clinical Mtb strains, while no cross-resistance to conventional antituberculosis drugs was observed. This suggested a novel mechanism of action, which was confirmed by chemoproteomic analysis leading to the identification of BCG_3193 and BCG_3827 as putative targets of the series with unknown function. Initial structure–activity relationship studies have resulted in compounds with moderate to potent antitubercular activity and improved physicochemical properties.
Collapse
Affiliation(s)
- Colin R Wilson
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Richard K Gessner
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Atica Moosa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town , Rondebosch 7701, South Africa
| | - Ronnett Seldon
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa
| | - Valerie Mizrahi
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa
| | - Candice Soares de Melo
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Sandile B Simelane
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Aloysius Nchinda
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Efrem Abay
- Department of Medicine, Division of Clinical Pharmacology, Drug Discovery and Development Centre (H3D), University of Cape Town , Observatory, 7925, South Africa
| | - Dale Taylor
- Department of Medicine, Division of Clinical Pharmacology, Drug Discovery and Development Centre (H3D), University of Cape Town , Observatory, 7925, South Africa
| | - Mathew Njoroge
- Department of Medicine, Division of Clinical Pharmacology, Drug Discovery and Development Centre (H3D), University of Cape Town , Observatory, 7925, South Africa
| | - Christel Brunschwig
- Department of Medicine, Division of Clinical Pharmacology, Drug Discovery and Development Centre (H3D), University of Cape Town , Observatory, 7925, South Africa
| | - Nina Lawrence
- Department of Medicine, Division of Clinical Pharmacology, Drug Discovery and Development Centre (H3D), University of Cape Town , Observatory, 7925, South Africa
| | - Helena I M Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | - Frederick A Sirgel
- DST/NRF Centre of Excellence for Biomedical TB Research, SA MRC Centre for TB Research, Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University , 7505 Tygerberg, South Africa
| | - Paul van Helden
- DST/NRF Centre of Excellence for Biomedical TB Research, SA MRC Centre for TB Research, Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University , 7505 Tygerberg, South Africa
| | - C John Harris
- CJH Consultants , Ford Cottage, South Weirs, Brockenhurst, Hampshire SO42 7UQ, U.K
| | - Richard Gordon
- Strategic Health Innovation Partnerships (SHIP), South African Medical Research Council , Parow Valley, Cape Town, South Africa
| | - Sonja Ghidelli-Disse
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline , Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Hannah Pflaumer
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline , Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Markus Boesche
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline , Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Gerard Drewes
- Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline , Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Olalla Sanz
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Gracia Santos
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Maria José Rebollo-Lopez
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Beatriz Urones
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Carolyn Selenski
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | | | - Matthew Axtman
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Joël Lelièvre
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Lluis Ballell
- Diseases of the Developing World, GlaxoSmithKline , Calle Severo Ochoa 2, 28760 Tres Cantos, Madrid, Spain
| | - Rudolf Mueller
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Leslie J Street
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Sandeep R Ghorpade
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, Drug Discovery and Development Centre (H3D), University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa.,South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| |
Collapse
|
76
|
Cole ST. Inhibiting Mycobacterium tuberculosis within and without. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0506. [PMID: 27672155 DOI: 10.1098/rstb.2015.0506] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2016] [Indexed: 01/05/2023] Open
Abstract
Tuberculosis remains a scourge of global health with shrinking treatment options due to the spread of drug-resistant strains of Mycobacterium tuberculosis Intensive efforts have been made in the past 15 years to find leads for drug development so that better, more potent drugs inhibiting new targets could be produced and thus shorten treatment duration. Initial attempts focused on repurposing drugs that had been developed for other therapeutic areas but these agents did not meet their goals in clinical trials. Attempts to find new lead compounds employing target-based screens were unsuccessful as the leads were inactive against M. tuberculosis Greater success was achieved using phenotypic screening against live tubercle bacilli and this gave rise to the drugs bedaquiline, pretomanid and delamanid, currently in phase III trials. Subsequent phenotypic screens also uncovered new leads and targets but several of these targets proved to be promiscuous and inhibited by a variety of seemingly unrelated pharmacophores. This setback sparked an interest in alternative screening approaches that mimic the disease state more accurately. Foremost among these were cell-based screens, often involving macrophages, as these should reflect the bacterium's niche in the host more faithfully. A major advantage of this approach is its ability to uncover functions that are central to infection but not necessarily required for growth in vitro For instance, inhibition of virulence functions mediated by the ESX-1 secretion system severely attenuates intracellular M. tuberculosis, preventing intercellular spread and ultimately limiting tissue damage. Cell-based screens have highlighted the druggability of energy production via the electron transport chain and cholesterol metabolism. Here, I review the scientific progress and the pipeline, but warn against over-optimism due to the lack of industrial commitment for tuberculosis drug development and other socio-economic factors.This article is part of the themed issue 'The new bacteriology'.
Collapse
Affiliation(s)
- Stewart T Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, Station 19, 1015 Lausanne, Switzerland
| |
Collapse
|
77
|
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]
|
78
|
Susceptibility of Mycobacterium tuberculosis Cytochrome bd Oxidase Mutants to Compounds Targeting the Terminal Respiratory Oxidase, Cytochrome c. Antimicrob Agents Chemother 2017; 61:AAC.01338-17. [PMID: 28760899 DOI: 10.1128/aac.01338-17] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/22/2017] [Indexed: 01/04/2023] Open
Abstract
We deleted subunits I (cydA) and II (cydB) of the Mycobacterium tuberculosis cytochrome bd menaquinol oxidase. The resulting ΔcydA and ΔcydAB mutants were hypersusceptible to compounds targeting the mycobacterial bc1 menaquinol-cytochrome c oxidoreductase and exhibited bioenergetic profiles indistinguishable from strains deficient in the ABC-type transporter, CydDC, predicted to be essential for cytochrome bd assembly. These results confirm CydAB and CydDC as potential targets for drugs aimed at inhibiting a terminal respiratory oxidase implicated in pathogenesis.
Collapse
|
79
|
Boot M, Jim KK, Liu T, Commandeur S, Lu P, Verboom T, Lill H, Bitter W, Bald D. A fluorescence-based reporter for monitoring expression of mycobacterial cytochrome bd in response to antibacterials and during infection. Sci Rep 2017; 7:10665. [PMID: 28878275 PMCID: PMC5587683 DOI: 10.1038/s41598-017-10944-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/16/2017] [Indexed: 12/16/2022] Open
Abstract
Cytochrome bd is a component of the oxidative phosphorylation pathway in many Gram-positive and Gram-negative bacteria. Next to its role as a terminal oxidase in the respiratory chain this enzyme plays an important role as a survival factor in the bacterial stress response. In Mycobacterium tuberculosis and related mycobacterial strains, cytochrome bd is an important component of the defense system against antibacterial drugs. In this report we describe and evaluate an mCherry-based fluorescent reporter for detection of cytochrome bd expression in Mycobacterium marinum. Cytochrome bd was induced by mycolic acid biosynthesis inhibitors such as isoniazid and most prominently by drugs targeting oxidative phosphorylation. We observed no induction by inhibitors of protein-, DNA- or RNA-synthesis. The constructed expression reporter was suitable for monitoring mycobacterial cytochrome bd expression during mouse macrophage infection and in a zebrafish embryo infection model when using Mycobacterium marinum. Interestingly, in both these infection models cytochrome bd levels were considerably higher than during in vitro culturing of M. marinum. The expression reporter described here can be a valuable tool for elucidating the role of cytochrome bd as a survival factor.
Collapse
Affiliation(s)
- Maikel Boot
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Kin Ki Jim
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Ting Liu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Susanna Commandeur
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Ping Lu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Theo Verboom
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Holger Lill
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.,Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Dirk Bald
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
| |
Collapse
|
80
|
Mukherjee R, Chandra Pal A, Banerjee M. Enabling faster Go/No-Go decisions through secondary screens in anti-mycobacterial drug discovery. Tuberculosis (Edinb) 2017; 106:44-52. [PMID: 28802404 DOI: 10.1016/j.tube.2017.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/30/2017] [Accepted: 06/28/2017] [Indexed: 10/19/2022]
Abstract
Management of tuberculosis, already a global health emergency, is becoming increasingly challenging with extensive misuse of second line drugs and their inaccessibility to eighty percent of the eligible patients. Rising statistics of antimicrobial resistance underscores the need for a set of completely new and more effective class of compounds with novel mechanisms of action that can be administered in combination to replace and shorten the present intensive six months regimen. In this review, we stress on the importance and the successes of phenotypic screening for discovery of anti-mycobacterial compound and discuss the importance of performing secondary screens and counter screens to get early estimate on compound's potentials for a successful development. We also highlight the recent advances and the related caveats in the assays that have been developed and discuss new screening modalities that can be incorporated during hit-selection to gain a quick insight into the mechanism of action, thus enabling quicker decisions in a hit triage.
Collapse
Affiliation(s)
- Raju Mukherjee
- Division of Biology, Indian Institute of Science Education and Research, Karakambadi Road, Tirupati, 517507, India.
| | - Anup Chandra Pal
- Division of Biology, Indian Institute of Science Education and Research, Karakambadi Road, Tirupati, 517507, India
| | - Mousumi Banerjee
- Indian Institute of Technology, Tirupati, Renigunta Road, Tirupati, 517506, India
| |
Collapse
|
81
|
Moraski GC, Bristol R, Seeger N, Boshoff HI, Tsang PSY, Miller MJ. Preparation and Evaluation of Potent Pentafluorosulfanyl-Substituted Anti-Tuberculosis Compounds. ChemMedChem 2017; 12:1108-1115. [PMID: 28654200 PMCID: PMC5603227 DOI: 10.1002/cmdc.201700170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/13/2017] [Indexed: 01/10/2023]
Abstract
The global fight to stop tuberculosis (TB) remains a great challenge, particularly with the increase in drug-resistant strains and a lack of funding to support the development of new treatments. To bolster a precarious drug pipeline, we prepared a focused panel of eight pentafluorosulfanyl (SF5 ) compounds which were screened for their activity against Mycobacterium tuberculosis (Mtb) H37Rv in three different assay conditions and media. All eight compounds had sub-micromolar potency, and four displayed MICs <100 nm. Seven compounds were evaluated against non-replicating and mono-drug-resistant Mtb, and for their ability to inhibit Mtb within the macrophage. The greatest potency was observed against intracellular Mtb (MIC <10 nm for three compounds), which is often the most challenging to target. In general, the SF5 -bearing compounds were very similar to their CF3 counterparts, with the major differences observed being their in vitro ADME properties. Two SF5 -bearing compounds were found to have greater protein binding than their corresponding CF3 counterparts, but were also less metabolized in human microsomes, resulting in longer half-lives.
Collapse
Affiliation(s)
- Garrett C Moraski
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Ryan Bristol
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Natalie Seeger
- Department of Chemistry and Biochemistry, 103 Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Helena I Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases (LCID), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 33 North Drive, Bethesda, MD, 20892, USA
| | - Patricia Siu-Yee Tsang
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases (LCID), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 33 North Drive, Bethesda, MD, 20892, USA
| | - Marvin J Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 347 Nieuwland Hall of Science, Notre Dame, IN, 46556, USA
| |
Collapse
|
82
|
Exploiting the synthetic lethality between terminal respiratory oxidases to kill Mycobacterium tuberculosis and clear host infection. Proc Natl Acad Sci U S A 2017; 114:7426-7431. [PMID: 28652330 DOI: 10.1073/pnas.1706139114] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The recent discovery of small molecules targeting the cytochrome bc1 :aa3 in Mycobacterium tuberculosis triggered interest in the terminal respiratory oxidases for antituberculosis drug development. The mycobacterial cytochrome bc1 :aa3 consists of a menaquinone:cytochrome c reductase (bc1 ) and a cytochrome aa3 -type oxidase. The clinical-stage drug candidate Q203 interferes with the function of the subunit b of the menaquinone:cytochrome c reductase. Despite the affinity of Q203 for the bc1 :aa3 complex, the drug is only bacteriostatic and does not kill drug-tolerant persisters. This raises the possibility that the alternate terminal bd-type oxidase (cytochrome bd oxidase) is capable of maintaining a membrane potential and menaquinol oxidation in the presence of Q203. Here, we show that the electron flow through the cytochrome bd oxidase is sufficient to maintain respiration and ATP synthesis at a level high enough to protect M. tuberculosis from Q203-induced bacterial death. Upon genetic deletion of the cytochrome bd oxidase-encoding genes cydAB, Q203 inhibited mycobacterial respiration completely, became bactericidal, killed drug-tolerant mycobacterial persisters, and rapidly cleared M. tuberculosis infection in vivo. These results indicate a synthetic lethal interaction between the two terminal respiratory oxidases that can be exploited for anti-TB drug development. Our findings should be considered in the clinical development of drugs targeting the cytochrome bc1 :aa3 , as well as for the development of a drug combination targeting oxidative phosphorylation in M. tuberculosis.
Collapse
|
83
|
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
|
84
|
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
|
85
|
Singh V, Donini S, Pacitto A, Sala C, Hartkoorn RC, Dhar N, Keri G, Ascher DB, Mondésert G, Vocat A, Lupien A, Sommer R, Vermet H, Lagrange S, Buechler J, Warner D, McKinney JD, Pato J, Cole ST, Blundell TL, Rizzi M, Mizrahi V. The Inosine Monophosphate Dehydrogenase, GuaB2, Is a Vulnerable New Bactericidal Drug Target for Tuberculosis. ACS Infect Dis 2017; 3:5-17. [PMID: 27726334 PMCID: PMC5241705 DOI: 10.1021/acsinfecdis.6b00102] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Indexed: 12/14/2022]
Abstract
VCC234718, a molecule with growth inhibitory activity against Mycobacterium tuberculosis (Mtb), was identified by phenotypic screening of a 15344-compound library. Sequencing of a VCC234718-resistant mutant identified a Y487C substitution in the inosine monophosphate dehydrogenase, GuaB2, which was subsequently validated to be the primary molecular target of VCC234718 in Mtb. VCC234718 inhibits Mtb GuaB2 with a Ki of 100 nM and is uncompetitive with respect to IMP and NAD+. This compound binds at the NAD+ site, after IMP has bound, and makes direct interactions with IMP; therefore, the inhibitor is by definition uncompetitive. VCC234718 forms strong pi interactions with the Y487 residue side chain from the adjacent protomer in the tetramer, explaining the resistance-conferring mutation. In addition to sensitizing Mtb to VCC234718, depletion of GuaB2 was bactericidal in Mtb in vitro and in macrophages. When supplied at a high concentration (≥125 μM), guanine alleviated the toxicity of VCC234718 treatment or GuaB2 depletion via purine salvage. However, transcriptional silencing of guaB2 prevented Mtb from establishing an infection in mice, confirming that Mtb has limited access to guanine in this animal model. Together, these data provide compelling validation of GuaB2 as a new tuberculosis drug target.
Collapse
Affiliation(s)
- Vinayak Singh
- MRC/NHLS/UCT Molecular Mycobacteriology
Research Unit & DST/NRF Centre of Excellence for Biomedical TB
Research, Institute of Infectious Disease and Molecular Medicine &
Department of Pathology, University of Cape
Town, Anzio Road, Observatory 7925, South Africa
| | - Stefano Donini
- Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio 6, 28100 Novara, Italy
| | - Angela Pacitto
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Claudia Sala
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Ruben C. Hartkoorn
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Neeraj Dhar
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Gyorgy Keri
- Vichem
Chemie, Herman Ottó
út 15, Budapest, 1022 Hungary
| | - David B. Ascher
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Guillaume Mondésert
- Sanofi-Aventis Research
& Development, Infectious Diseases Unit,
Biology Group, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280 Marcy L’Etoile, France
| | - Anthony Vocat
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Andréanne Lupien
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Raphael Sommer
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Hélène Vermet
- Sanofi-Aventis Research
& Development, Infectious Diseases Unit,
Biology Group, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280 Marcy L’Etoile, France
| | - Sophie Lagrange
- Sanofi-Aventis Research
& Development, Infectious Diseases Unit,
Biology Group, Campus Mérieux, 1541 avenue Marcel Mérieux, 69280 Marcy L’Etoile, France
| | - Joe Buechler
- Alere (San Diego), Summer Ridge Road, San Diego, California 92121, United States
| | - Digby
F. Warner
- MRC/NHLS/UCT Molecular Mycobacteriology
Research Unit & DST/NRF Centre of Excellence for Biomedical TB
Research, Institute of Infectious Disease and Molecular Medicine &
Department of Pathology, University of Cape
Town, Anzio Road, Observatory 7925, South Africa
| | - John D. McKinney
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Janos Pato
- Vichem
Chemie, Herman Ottó
út 15, Budapest, 1022 Hungary
| | - Stewart T. Cole
- Ecole Polytechnique Fédérale de Lausanne, Global Health Institute, Station 19, 1015 Lausanne, Switzerland
| | - Tom L. Blundell
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Menico Rizzi
- Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio 6, 28100 Novara, Italy
| | - Valerie Mizrahi
- MRC/NHLS/UCT Molecular Mycobacteriology
Research Unit & DST/NRF Centre of Excellence for Biomedical TB
Research, Institute of Infectious Disease and Molecular Medicine &
Department of Pathology, University of Cape
Town, Anzio Road, Observatory 7925, South Africa
| |
Collapse
|
86
|
Stringer T, Seldon R, Liu N, Warner DF, Tam C, Cheng LW, Land KM, Smith PJ, Chibale K, Smith GS. Antimicrobial activity of organometallic isonicotinyl and pyrazinyl ferrocenyl-derived complexes. Dalton Trans 2017; 46:9875-9885. [DOI: 10.1039/c7dt01952a] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Isonicotinyl and pyrazinyl ferrocenyl-derived complexes were evaluatedin vitrofor antimycobacterial and antiparasitic activity.
Collapse
|
87
|
Recent advancements in the development of anti-tuberculosis drugs. Bioorg Med Chem Lett 2016; 27:370-386. [PMID: 28017531 DOI: 10.1016/j.bmcl.2016.11.084] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/16/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023]
Abstract
Modern chemotherapy has significantly improved patient outcomes against drug-sensitive tuberculosis. However, the rapid emergence of drug-resistant tuberculosis, together with the bacterium's ability to persist and remain latent present a major public health challenge. To overcome this problem, research into novel anti-tuberculosis targets and drug candidates is thus of paramount importance. This review article provides an overview of tuberculosis highlighting the recent advances and tools that are employed in the field of anti-tuberculosis drug discovery. The predominant focus is on anti-tuberculosis agents that are currently in the pipeline, i.e. clinical trials.
Collapse
|
88
|
Bioluminescent Reporters for Rapid Mechanism of Action Assessment in Tuberculosis Drug Discovery. Antimicrob Agents Chemother 2016; 60:6748-6757. [PMID: 27572410 DOI: 10.1128/aac.01178-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/26/2016] [Indexed: 12/12/2022] Open
Abstract
The tuberculosis (TB) drug discovery pipeline is fueled by compounds identified in whole-cell screens against the causative agent, Mycobacterium tuberculosis Phenotypic screening enables the selection of molecules that inhibit essential cellular functions in live, intact bacilli grown under a chosen in vitro condition. However, deducing the mechanism of action (MOA), which is important to avoid promiscuous targets, often requires significant biological resources in a lengthy process that risks decoupling medicinal chemistry and biology efforts. Therefore, there is a need to develop methods enabling rapid MOA assessment of putative "actives" for triage decisions. Here, we describe a modified version of a bioluminescence reporter assay that allows nondestructive detection of compounds targeting either of two macromolecular processes in M. tuberculosis: cell wall biosynthesis or maintenance of DNA integrity. Coupling the luxCDABE operon from Photorhabdus luminescens to mycobacterial promoters driving expression of the iniBAC operon (PiniB-LUX) or the DNA damage-inducible genes, recA (PrecA-LUX) or radA (PradA-LUX), provided quantitative detection in real time of compounds triggering expression of any of these promoters over an extended 10- to 12-day incubation. Testing against known anti-TB agents confirmed the specificity of each reporter in registering the MOA of the applied antibiotic in M. tuberculosis, independent of bactericidal or bacteriostatic activity. Moreover, profiles obtained for experimental compounds indicated the potential to infer complex MOAs in which multiple cellular processes are disrupted. These results demonstrate the utility of the reporters for early triage of compounds based on the provisional MOA and suggest their application to investigate polypharmacology in known and experimental anti-TB agents.
Collapse
|
89
|
Phummarin N, Boshoff HI, Tsang PS, Dalton J, Wiles S, Barry Rd CE, Copp BR. SAR and identification of 2-(quinolin-4-yloxy)acetamides as Mycobacterium tuberculosis cytochrome bc1 inhibitors. MEDCHEMCOMM 2016; 7:2122-2127. [PMID: 28337336 PMCID: PMC5292992 DOI: 10.1039/c6md00236f] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/17/2016] [Indexed: 12/13/2022]
Abstract
New antimycobacterial 2-(quinoline-4-yloxy)acetamides were prepared, and using gene deletion and resistant mutants, we conclude that the compound class inhibits the mycobacterial cytochrome bc1 complex.
A previous phenotypic screen by GSK identified 2-(quinolin-4-yloxy)acetamides as potent growth inhibitors of Mycobacterium tuberculosis (Mtb). We report the results of a preliminary structure–activity relationship (SAR) study of the compound class which has yielded more potent inhibitors. An Mtb cytochrome bd oxidase deletion mutant (cydKO) was found to be hypersensitive to most members of the compound library, while strains carrying single-nucleotide polymorphisms of the qcrB gene, which encodes a subunit of the menaquinol cytochrome c oxidoreductase (bc1) complex, were resistant to the library. These results identify that the 2-(quinolin-4-yloxy)acetamide class of Mtb growth inhibitors can be added to the growing number of scaffolds that target the M. tuberculosis bc1 complex.
Collapse
Affiliation(s)
- Narisa Phummarin
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand . ; ; Tel: +64 9 3737599
| | - Helena I Boshoff
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - Patricia S Tsang
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - James Dalton
- Bioluminescent Superbugs Lab , Department of Molecular Medicine and Pathology , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab , Department of Molecular Medicine and Pathology , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand ; Te Pūnaha Matatini , c/o University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand
| | - Clifton E Barry Rd
- Tuberculosis Research Section , Laboratory of Clinical Infectious Diseases , National Institute of Allergy and Infectious Disease , National Institutes of Health , Bethesda , Maryland , USA
| | - Brent R Copp
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand . ; ; Tel: +64 9 3737599
| |
Collapse
|
90
|
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
|
91
|
Chinta KC, Saini V, Glasgow JN, Mazorodze JH, Rahman MA, Reddy D, Lancaster JR, Steyn AJC. The emerging role of gasotransmitters in the pathogenesis of tuberculosis. Nitric Oxide 2016; 59:28-41. [PMID: 27387335 DOI: 10.1016/j.niox.2016.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 06/30/2016] [Indexed: 12/17/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is a facultative intracellular pathogen and the second largest contributor to global mortality caused by an infectious agent after HIV. In infected host cells, Mtb is faced with a harsh intracellular environment including hypoxia and the release of nitric oxide (NO) and carbon monoxide (CO) by immune cells. Hypoxia, NO and CO induce a state of in vitro dormancy where Mtb senses these gases via the DosS and DosT heme sensor kinase proteins, which in turn induce a set of ∼47 genes, known as the Mtb Dos dormancy regulon. On the contrary, both iNOS and HO-1, which produce NO and CO, respectively, have been shown to be important against mycobacterial disease progression. In this review, we discuss the impact of O2, NO and CO on Mtb physiology and in host responses to Mtb infection as well as the potential role of another major endogenous gas, hydrogen sulfide (H2S), in Mtb pathogenesis.
Collapse
Affiliation(s)
- Krishna C Chinta
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vikram Saini
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA; UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joel N Glasgow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James H Mazorodze
- KwaZulu-Natal Research Institute for TB and HIV (KRITH), Durban, South Africa
| | - Md Aejazur Rahman
- KwaZulu-Natal Research Institute for TB and HIV (KRITH), Durban, South Africa
| | - Darshan Reddy
- Department of Cardiothoracic Surgery, Nelson R Mandela School of Medicine, University of KwaZulu Natal, Durban, South Africa
| | - Jack R Lancaster
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrie J C Steyn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA; KwaZulu-Natal Research Institute for TB and HIV (KRITH), Durban, South Africa; UAB Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
92
|
Moraski GC, Seeger N, Miller PA, Oliver AG, Boshoff HI, Cho S, Mulugeta S, Anderson JR, Franzblau SG, Miller MJ. Arrival of Imidazo[2,1-b]thiazole-5-carboxamides: Potent Anti-tuberculosis Agents That Target QcrB. ACS Infect Dis 2016; 2:393-8. [PMID: 27627627 DOI: 10.1021/acsinfecdis.5b00154] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Increasing interest in the potent anti-tuberculosis activity and the novel target (QcrB) of imidazo[1,2-a]pyridine-3-carboxamides encouraged extended structure-activity relationship studies of additional scaffolds. This study reports on the in vitro profiling of the imidazo[2,1-b]thiazole-5-carboxamides as a new promising class of anti-tuberculosis compounds endowed with nanomolar potency against replicating and drug-resistant Mycobacterium tuberculosis (Mtb) as well as low toxicity to VERO cells. Compounds 6, 16, and 17 had MIC values <10 nM and toxicity >100 μM. On-target selectivity of this series was confirmed by cross-resistance of specific QcrB mutants as well as the hypersusceptibility of a mutant with a functional gene deletion of the alternative cytochrome bd oxidase. Additionally, to demonstrate selectivity, three analogues (6, 15, 17) were broadly screened against a diverse set of eight strains of bacteria, including both Gram-positive and Gram-negative as well as six disease-causing non-tuberculosis mycobacteria. Finally, compounds 16 and 17 were found to be active in macrophages infected with Mtb.
Collapse
Affiliation(s)
- Garrett C. Moraski
- Department of Chemistry and Biochemistry, Montana State University, 103 Chemistry and Biochemistry Building, Bozeman, Montana 59717, United States
| | - Natalie Seeger
- Department of Chemistry and Biochemistry, Montana State University, 103 Chemistry and Biochemistry Building, Bozeman, Montana 59717, United States
| | - Patricia A. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Allen G. Oliver
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| | - Helena I. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical
Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Sanghyun Cho
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Surafel Mulugeta
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Jeffery R. Anderson
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Marvin J. Miller
- Department of Chemistry and Biochemistry, University of Notre Dame, 251 Nieuwland Science Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
93
|
Pienaar E, Matern WM, Linderman JJ, Bader JS, Kirschner DE. Multiscale Model of Mycobacterium tuberculosis Infection Maps Metabolite and Gene Perturbations to Granuloma Sterilization Predictions. Infect Immun 2016; 84:1650-1669. [PMID: 26975995 PMCID: PMC4862722 DOI: 10.1128/iai.01438-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/08/2016] [Indexed: 02/06/2023] Open
Abstract
Granulomas are a hallmark of tuberculosis. Inside granulomas, the pathogen Mycobacterium tuberculosis may enter a metabolically inactive state that is less susceptible to antibiotics. Understanding M. tuberculosis metabolism within granulomas could contribute to reducing the lengthy treatment required for tuberculosis and provide additional targets for new drugs. Two key adaptations of M. tuberculosis are a nonreplicating phenotype and accumulation of lipid inclusions in response to hypoxic conditions. To explore how these adaptations influence granuloma-scale outcomes in vivo, we present a multiscale in silico model of granuloma formation in tuberculosis. The model comprises host immunity, M. tuberculosis metabolism, M. tuberculosis growth adaptation to hypoxia, and nutrient diffusion. We calibrated our model to in vivo data from nonhuman primates and rabbits and apply the model to predict M. tuberculosis population dynamics and heterogeneity within granulomas. We found that bacterial populations are highly dynamic throughout infection in response to changing oxygen levels and host immunity pressures. Our results indicate that a nonreplicating phenotype, but not lipid inclusion formation, is important for long-term M. tuberculosis survival in granulomas. We used virtual M. tuberculosis knockouts to predict the impact of both metabolic enzyme inhibitors and metabolic pathways exploited to overcome inhibition. Results indicate that knockouts whose growth rates are below ∼66% of the wild-type growth rate in a culture medium featuring lipid as the only carbon source are unable to sustain infections in granulomas. By mapping metabolite- and gene-scale perturbations to granuloma-scale outcomes and predicting mechanisms of sterilization, our method provides a powerful tool for hypothesis testing and guiding experimental searches for novel antituberculosis interventions.
Collapse
Affiliation(s)
- Elsje Pienaar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William M Matern
- Department of Biomedical Engineering and High-Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Joel S Bader
- Department of Biomedical Engineering and High-Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
94
|
van der Westhuyzen R, Winks S, Wilson CR, Boyle GA, Gessner RK, Soares de Melo C, Taylor D, de Kock C, Njoroge M, Brunschwig C, Lawrence N, Rao SPS, Sirgel F, van Helden P, Seldon R, Moosa A, Warner DF, Arista L, Manjunatha UH, Smith PW, Street LJ, Chibale K. Pyrrolo[3,4-c]pyridine-1,3(2H)-diones: A Novel Antimycobacterial Class Targeting Mycobacterial Respiration. J Med Chem 2015; 58:9371-81. [PMID: 26551248 DOI: 10.1021/acs.jmedchem.5b01542] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
High-throughput screening of a library of small polar molecules against Mycobacterium tuberculosis led to the identification of a phthalimide-containing ester hit compound (1), which was optimized for metabolic stability by replacing the ester moiety with a methyl oxadiazole bioisostere. A route utilizing polymer-supported reagents was designed and executed to explore structure-activity relationships with respect to the N-benzyl substituent, leading to compounds with nanomolar activity. The frontrunner compound (5h) from these studies was well tolerated in mice. A M. tuberculosis cytochrome bd oxidase deletion mutant (ΔcydKO) was hyper-susceptible to compounds from this series, and a strain carrying a single point mutation in qcrB, the gene encoding a subunit of the menaquinol cytochrome c oxidoreductase, was resistant to compounds in this series. In combination, these observations indicate that this novel class of antimycobacterial compounds inhibits the cytochrome bc1 complex, a validated drug target in M. tuberculosis.
Collapse
Affiliation(s)
- Renier van der Westhuyzen
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Susan Winks
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Colin R Wilson
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Grant A Boyle
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Richard K Gessner
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Candice Soares de Melo
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Dale Taylor
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town , Observatory 7925, South Africa
| | - Carmen de Kock
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town , Observatory 7925, South Africa
| | - Mathew Njoroge
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Christel Brunschwig
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Nina Lawrence
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Srinivasa P S Rao
- Novartis Institute for Tropical Diseases (NITD) , 10 Biopolis Road, Chromos no. 05-01, Singapore 138670, Singapore
| | - Frederick Sirgel
- DST/NRF Centre of Excellence in Biomedical TB Research, SA MRC Centre for TB Research, Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University , Tygerberg 7505, South Africa
| | - Paul van Helden
- DST/NRF Centre of Excellence in Biomedical TB Research, SA MRC Centre for TB Research, Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University , Tygerberg 7505, South Africa
| | - Ronnett Seldon
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Atica Moosa
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town , Rondebosch 7701, South Africa
| | - Digby F Warner
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa
| | - Luca Arista
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research , Novartis Campus, CH-4056 Basel, Switzerland
| | - Ujjini H Manjunatha
- Novartis Institute for Tropical Diseases (NITD) , 10 Biopolis Road, Chromos no. 05-01, Singapore 138670, Singapore
| | - Paul W Smith
- Novartis Institute for Tropical Diseases (NITD) , 10 Biopolis Road, Chromos no. 05-01, Singapore 138670, Singapore
| | - Leslie J Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa.,South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town , Rondebosch 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Rondebosch 7701, South Africa
| |
Collapse
|
95
|
Soares de Melo C, Candice SDM, Feng TS, van der Westhuyzen R, Gessner RK, Street LJ, Morgans GL, Warner DF, Moosa A, Naran K, Lawrence N, Boshoff HIM, Barry CE, Harris CJ, Gordon R, Chibale K. Aminopyrazolo[1,5-a]pyrimidines as potential inhibitors of Mycobacterium tuberculosis: Structure activity relationships and ADME characterization. Bioorg Med Chem 2015; 23:7240-50. [PMID: 26522089 DOI: 10.1016/j.bmc.2015.10.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/05/2015] [Accepted: 10/13/2015] [Indexed: 11/24/2022]
Abstract
Whole-cell high-throughput screening of a diverse SoftFocus library against Mycobacterium tuberculosis (Mtb) generated a novel aminopyrazolo[1,5-a]pyrimidine hit series. The synthesis and structure activity relationship studies identified compounds with potent antimycobacterial activity. The SAR of over 140 compounds shows that the 2-pyridylmethylamine moiety at the C-7 position of the pyrazolopyrimidine scaffold was important for Mtb activity, whereas the C-3 position offered a higher degree of flexibility. The series was also profiled for in vitro cytotoxicity and microsomal metabolic stability as well as physicochemical properties. Consequently liabilities to be addressed in a future lead optimization campaign have been identified.
Collapse
Affiliation(s)
| | - Soares de Melo Candice
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Tzu-Shean Feng
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Renier van der Westhuyzen
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Richard K Gessner
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Leslie J Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Digby F Warner
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Atica Moosa
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Krupa Naran
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Nina Lawrence
- Division of Clinical Pharmacology, University of Cape Town, Rondebosch 7701, South Africa
| | - Helena I M Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States
| | - C John Harris
- Cjh Consultants (Pharmaceutical R&D), Lydith, High Street, Eynsford, Kent DA4 0AB, United Kingdom
| | - Richard Gordon
- Strategic Health Innovation Partnerships (SHIP), South African Medical Research Council, Parow Valley, South Africa
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa.
| |
Collapse
|
96
|
Rybniker J, Vocat A, Sala C, Busso P, Pojer F, Benjak A, Cole ST. Lansoprazole is an antituberculous prodrug targeting cytochrome bc1. Nat Commun 2015; 6:7659. [PMID: 26158909 PMCID: PMC4510652 DOI: 10.1038/ncomms8659] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 05/29/2015] [Indexed: 12/28/2022] Open
Abstract
Better antibiotics capable of killing multi-drug-resistant Mycobacterium tuberculosis are urgently needed. Despite extensive drug discovery efforts, only a few promising candidates are on the horizon and alternative screening protocols are required. Here, by testing a panel of FDA-approved drugs in a host cell-based assay, we show that the blockbuster drug lansoprazole (Prevacid), a gastric proton-pump inhibitor, has intracellular activity against M. tuberculosis. Ex vivo pharmacokinetics and target identification studies reveal that lansoprazole kills M. tuberculosis by targeting its cytochrome bc1 complex through intracellular sulfoxide reduction to lansoprazole sulfide. This novel class of cytochrome bc1 inhibitors is highly active against drug-resistant clinical isolates and spares the human H+K+-ATPase thus providing excellent opportunities for targeting the major pathogen M. tuberculosis. Our finding provides proof of concept for hit expansion by metabolic activation, a powerful tool for antibiotic screens. Tuberculosis control is threatened by the continued emergence of drug-resistant strains. Here, Rybniker et al. screen a library of FDA-approved drugs and identify a gastric proton pump inhibitor that also has antituberculosis activity and targets the bacterial cytochrome bc1 complex.
Collapse
Affiliation(s)
- Jan Rybniker
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.,1st Department of Internal Medicine, University of Cologne, D-50937 Cologne, Germany
| | - Anthony Vocat
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Claudia Sala
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Philippe Busso
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Florence Pojer
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Andrej Benjak
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Stewart T Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
97
|
Lu P, Heineke MH, Koul A, Andries K, Cook GM, Lill H, van Spanning R, Bald D. The cytochrome bd-type quinol oxidase is important for survival of Mycobacterium smegmatis under peroxide and antibiotic-induced stress. Sci Rep 2015; 5:10333. [PMID: 26015371 PMCID: PMC4450806 DOI: 10.1038/srep10333] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/09/2015] [Indexed: 12/22/2022] Open
Abstract
Targeting respiration and ATP synthesis has received strong interest as a new
strategy for combatting drug-resistant Mycobacterium tuberculosis.
Mycobacteria employ a respiratory chain terminating with two branches. One of the
branches includes a cytochrome bc1 complex and an
aa3-type cytochrome c oxidase while the other branch
terminates with a cytochrome bd-type quinol oxidase. In this communication we
show that genetic inactivation of cytochrome bd, but not of cytochrome
bc1, enhances the susceptibility of Mycobacterium
smegmatis to hydrogen peroxide and antibiotic-induced stress. The type-II
NADH dehydrogenase effector clofazimine and the ATP synthase inhibitor bedaquiline
were bacteriostatic against wild-type M. smegmatis, but strongly bactericidal
against a cytochrome bd mutant. We also demonstrated that the quinone-analog
aurachin D inhibited mycobacterial cytochrome bd at sub-micromolar
concentrations. Our results identify cytochrome bd as a key survival factor
in M. smegmatis during antibiotic stress. Targeting the cytochrome bd
respiratory branch therefore appears to be a promising strategy that may enhance the
bactericidal activity of existing tuberculosis drugs.
Collapse
Affiliation(s)
- Ping Lu
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Marieke H Heineke
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Anil Koul
- Infectious diseases and vaccines therapeutic area, Janssen Research &Development, Johnson &Johnson Pharmaceuticals, Turnhoutseweg 30, 2340-Beerse, Belgium
| | - Koen Andries
- Infectious diseases and vaccines therapeutic area, Janssen Research &Development, Johnson &Johnson Pharmaceuticals, Turnhoutseweg 30, 2340-Beerse, Belgium
| | - Gregory M Cook
- Department of Microbiology and Immunology, Otago School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Holger Lill
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Rob van Spanning
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Dirk Bald
- Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Earth- and Life Sciences, VU University Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
98
|
Borisov VB, Forte E, Siletsky SA, Arese M, Davletshin AI, Sarti P, Giuffrè A. Cytochrome bd protects bacteria against oxidative and nitrosative stress: A potential target for next-generation antimicrobial agents. BIOCHEMISTRY (MOSCOW) 2015; 80:565-75. [DOI: 10.1134/s0006297915050077] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
99
|
Kim MS, Jang J, Ab Rahman NB, Pethe K, Berry EA, Huang LS. Isolation and Characterization of a Hybrid Respiratory Supercomplex Consisting of Mycobacterium tuberculosis Cytochrome bcc and Mycobacterium smegmatis Cytochrome aa3. J Biol Chem 2015; 290:14350-60. [PMID: 25861988 PMCID: PMC4505504 DOI: 10.1074/jbc.m114.624312] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Indexed: 11/06/2022] Open
Abstract
Recently, energy production pathways have been shown to be viable antitubercular drug targets to combat multidrug-resistant tuberculosis and eliminate pathogen in the dormant state. One family of drugs currently under development, the imidazo[1,2-a]pyridine derivatives, is believed to target the pathogen's homolog of the mitochondrial bc1 complex. This complex, denoted cytochrome bcc, is highly divergent from mitochondrial Complex III both in subunit structure and inhibitor sensitivity, making it a good target for drug development. There is no soluble cytochrome c in mycobacteria to transport electrons from the bcc complex to cytochrome oxidase. Instead, the bcc complex exists in a "supercomplex" with a cytochrome aa3-type cytochrome oxidase, presumably allowing direct electron transfer. We describe here purification and initial characterization of the mycobacterial cytochrome bcc-aa3 supercomplex using a strain of M. smegmatis that has been engineered to express the M. tuberculosis cytochrome bcc. The resulting hybrid supercomplex is stable during extraction and purification in the presence of dodecyl maltoside detergent. It is hoped that this purification procedure will potentiate functional studies of the complex as well as crystallographic studies of drug binding and provide structural insight into a third class of the bc complex superfamily.
Collapse
Affiliation(s)
- Mi-Sun Kim
- From the State University of New York Upstate Medical University, Syracuse, New York 13210
| | - Jichan Jang
- the Institut Pasteur Korea, Sampyeong-dong, Seongnam-si, Gyeonggi-do 463-400, Korea, and
| | - Nurlilah Binte Ab Rahman
- the Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Kevin Pethe
- the Institut Pasteur Korea, Sampyeong-dong, Seongnam-si, Gyeonggi-do 463-400, Korea, and the Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Edward A Berry
- From the State University of New York Upstate Medical University, Syracuse, New York 13210,
| | - Li-Shar Huang
- From the State University of New York Upstate Medical University, Syracuse, New York 13210,
| |
Collapse
|
100
|
Ochoa-Montaño B, Mohan N, Blundell TL. CHOPIN: a web resource for the structural and functional proteome of Mycobacterium tuberculosis. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav026. [PMID: 25833954 PMCID: PMC4381106 DOI: 10.1093/database/bav026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/01/2015] [Indexed: 11/18/2022]
Abstract
Tuberculosis kills more than a million people annually and presents increasingly high levels of resistance against current first line drugs. Structural information about Mycobacterium tuberculosis (Mtb) proteins is a valuable asset for the development of novel drugs and for understanding the biology of the bacterium; however, only about 10% of the ∼4000 proteins have had their structures determined experimentally. The CHOPIN database assigns structural domains and generates homology models for 2911 sequences, corresponding to ∼73% of the proteome. A sophisticated pipeline allows multiple models to be created using conformational states characteristic of different oligomeric states and ligand binding, such that the models reflect various functional states of the proteins. Additionally, CHOPIN includes structural analyses of mutations potentially associated with drug resistance. Results are made available at the web interface, which also serves as an automatically updated repository of all published Mtb experimental structures. Its RESTful interface allows direct and flexible access to structures and metadata via intuitive URLs, enabling easy programmatic use of the models. Database URL: http://structure.bioc.cam.ac.uk/chopin
Collapse
Affiliation(s)
- Bernardo Ochoa-Montaño
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge CB2 1GA, UK and Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Nishita Mohan
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge CB2 1GA, UK and Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge CB2 1GA, UK and Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge CB2 1GA, UK and Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| |
Collapse
|