1
|
Ahmad A, Akhtar J, Ahmad M, Islam A, Badruddeen, Khan MI, Khan MV, Owais M. Innovative Bedaquiline-Based Delivery Systems for Multidrug-Resistant Tuberculosis Treatment. Infect Disord Drug Targets 2025; 25:e18715265318306. [PMID: 39301910 DOI: 10.2174/0118715265318306240816104553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 09/22/2024]
Abstract
The increasing incidence of multidrug-resistant tuberculosis (MDR-TB) is one of the most challenging tasks in tuberculosis treatment. Conventional TB treatment regimens have proven ineffective in treating MDR-TB, thus demanding the development of new drugs followed by delivery systems. Bedaquiline, a novel anti-TB drug, has been reported to inhibit the ATP synthase required for the growth and replication of TB bacteria. Bedaquiline is able to target the persistent or latent form of TB, which remains difficult to treat with conventional drugs. This makes bedaquiline an important drug in the fight against MDR-TB. The drug has been approved by the US FDA as well as European Medicines Agency and is now widely used as part of combination therapy for the treatment of MDR-TB. Bedaquiline and its advanced drug delivery system play a key role in tackling MDR-TB, providing a much-needed boost to control and eventually eliminate the disease. However, the cost of the drug remains a concern, and efforts are underway to make bedaquiline more accessible and affordable to patients in resource-limited settings. Nevertheless, the development of bedaquiline nanoformulations represents a significant step forward in the fight against TB and offers hope to millions of patients across the globe.
Collapse
Affiliation(s)
- Asad Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Juber Akhtar
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Mohammad Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Badruddeen
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Mohammad Irfan Khan
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Mohsin Vahid Khan
- Department of Biosciences, Integral University, Dasauli Kursi Road, Lucknow, 226026, India
| | - Mohammad Owais
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
2
|
Basrai A, Blundell TL, Pandurangan AP. Computational analyses of drug resistance mutations in katG and emb complexes in Mycobacterium tuberculosis. Proteins 2025; 93:359-371. [PMID: 38483037 PMCID: PMC11623437 DOI: 10.1002/prot.26684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 12/07/2024]
Abstract
The number of antibiotic resistant pathogens is increasing rapidly, and with this comes a substantial socioeconomic cost that threatens much of the world. To alleviate this problem, we must use antibiotics in a more responsible and informed way, further our understanding of the molecular basis of drug resistance, and design new antibiotics. Here, we focus on a key drug-resistant pathogen, Mycobacterium tuberculosis, and computationally analyze trends in drug-resistant mutations in genes of the proteins embA, embB, embC, and katG, which play essential roles in the action of the first-line drugs ethambutol and isoniazid. We use docking to predict binding modes of isoniazid to katG that agree with suggested binding sites found in our laboratory using cryo-EM. Using mutant stability predictions, we recapitulate the idea that resistance occurs when katG's heme cofactor is destabilized rather than due to a decrease in affinity to isoniazid. Conversely, we have identified resistance mutations that affect the affinity of ethambutol more drastically than the affinity of the natural substrate of embB. With this, we illustrate that we can distinguish between the two types of drug resistance-cofactor destabilization and drug affinity reduction-suggesting potential uses in the prediction of novel drug-resistant mutations.
Collapse
Affiliation(s)
- Aadam Basrai
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Department of MedicineUniversity of CambridgeCambridgeUK
| | - Tom L. Blundell
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Department of MedicineUniversity of CambridgeCambridgeUK
| | - Arun Prasad Pandurangan
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Department of MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
3
|
Li J, Duan R, Traore ES, Nguyen RC, Davis I, Griffth WP, Goodwin DC, Jarzecki AA, Liu A. Indole N-Linked Hydroperoxyl Adduct of Protein-Derived Cofactor Modulating Catalase-Peroxidase Functions. Angew Chem Int Ed Engl 2024; 63:e202407018. [PMID: 39300819 DOI: 10.1002/anie.202407018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 09/22/2024]
Abstract
Bifunctional catalase-peroxidase (KatG) features a posttranslational methionine-tyrosine-tryptophan (MYW) crosslinked cofactor crucial for its catalase function, enabling pathogens to neutralize hydrogen peroxide during infection. We discovered the presence of indole nitrogen-linked hydroperoxyl adduct (MYW-OOH) in Mycobacterium tuberculosis KatG in the solution state under ambient conditions, suggesting its natural occurrence. By isolating predominantly MYW-OOH-containing KatG protein, we investigated the chemical stability and functional impact of MYW-OOH. We discovered that MYW-OOH inhibits catalase activity, presenting a unique temporary lock. Exposure to peroxide or increased temperature removes the hydroperoxyl adduct from the protein cofactor, converting MYW-OOH to MYW and restoring the detoxifying ability of the enzyme against hydrogen peroxide. Thus, the N-linked hydroperoxyl group is releasable. KatG with MYW-OOH represents a catalase dormant, but primed, state of the enzyme. These findings provide insight into chemical strategies targeting the bifunctional enzyme KatG in pathogens, highlighting the role of N-linked hydroperoxyl modifications in enzymatic function.
Collapse
Affiliation(s)
- Jiasong Li
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Ran Duan
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Ephrahime S Traore
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Romie C Nguyen
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Ian Davis
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Wendell P Griffth
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| | - Douglas C Goodwin
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA
| | - Andrzej A Jarzecki
- Department of Chemistry and Biochemistry, Brooklyn College, New York, NY 11210, USA
| | - Aimin Liu
- Department of Chemistry, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX 78249, USA
| |
Collapse
|
4
|
Liu A. Catalase-peroxidase (KatG): a potential frontier in tuberculosis drug development. Crit Rev Biochem Mol Biol 2024; 59:434-446. [PMID: 40013498 PMCID: PMC11935562 DOI: 10.1080/10409238.2025.2470630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 02/28/2025]
Abstract
Mycobacterium tuberculosis (Mtb) depends on the bifunctional enzyme catalase-peroxidase (KatG) for survival within the host. KatG exhibits both catalase and peroxidase activities, serving distinct yet critical roles. While its peroxidase activity is essential for activating the frontline tuberculosis drug isoniazid, its catalase activity protects Mtb from oxidative stress. This bifunctional enzyme is equipped with a unique, protein-derived cofactor, methionine-tyrosine-tryptophan (MYW), which enables catalase activity to efficiently disproportionate hydrogen peroxide in phagocytes. Recent studies reveal that the MYW cofactor naturally exists in a hydroperoxylated form (MYW-OOH) when cell cultures are grown under ambient conditions. New findings highlight a dynamic regulation of KatG activity, wherein the modification of the protein cofactor is removable-from MYW-OOH to MYW-at body temperature or in the presence of micromolar concentrations of hydrogen peroxide. This reversible modification modulates KatG's dual activities: MYW-OOH inhibits catalase activity while enhancing peroxidase activity, demonstrating the chemical accessibility of the cofactor. Such duality positions KatG as a unique target for tuberculosis drug development. Therapeutic strategies that exploit cofactor modification could hold promise, particularly against drug-resistant strains with impaired peroxidase activity. By selectively inhibiting catalase activity, these approaches would render Mtb more vulnerable to oxidative stress while enhancing isoniazid activation-a double-edged strategy for combating tuberculosis.
Collapse
Affiliation(s)
- Aimin Liu
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78256, USA
| |
Collapse
|
5
|
Perez RL, Chase J, Tanner R. Shared challenges to the control of complex intracellular neglected pathogens. Front Public Health 2024; 12:1423420. [PMID: 39324165 PMCID: PMC11422159 DOI: 10.3389/fpubh.2024.1423420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/13/2024] [Indexed: 09/27/2024] Open
Abstract
The complex intracellular pathogens Mycobacterium tuberculosis, Mycobacterium leprae, Leishmania spp., and Burkholderia pseudomallei, which cause tuberculosis, leprosy, leishmaniasis, and melioidosis respectively, represent major health threats with a significant global burden concentrated in low- and middle-income countries. While these diseases vary in their aetiology, pathology and epidemiology, they share key similarities in the biological and sociodemographic factors influencing their incidence and impact worldwide. In particular, their occurrence in resource-limited settings has important implications for research and development, disease prevalence and associated risk factors, as well as access to diagnostics and therapeutics. In accordance with the vision of the VALIDATE (VAccine deveLopment for complex Intracellular neglecteD pAThogeEns) Network, we consider shared challenges to the effective prevention, diagnosis and treatment of these diseases as shaped by both biological and social factors, illustrating the importance of taking an interdisciplinary approach. We further highlight how a cross-pathogen perspective may provide valuable insights for understanding and addressing challenges to the control of all four pathogens.
Collapse
Affiliation(s)
- Rebecca Lynn Perez
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- Wadham College, University of Oxford, Oxford, United Kingdom
| | - Jemima Chase
- Wadham College, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Wadham College, University of Oxford, Oxford, United Kingdom
- Department of Biology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Sultana M, Alam MM, Mistri SK, Mostafa Kamal SM, Ahsan CR, Yasmin M. Multi-drug resistant gene mutation analysis in Mycobacterium tuberculosis by molecular techniques. IRANIAN JOURNAL OF MICROBIOLOGY 2024; 16:459-469. [PMID: 39267928 PMCID: PMC11389769 DOI: 10.18502/ijm.v16i4.16304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Background and Objectives Rifampicin (RIF) and isoniazid (INH), two most potent antibiotics, are prescribed to cure tuberculosis. Mycobacterium tuberculosis, the causative agent of multidrug-resistant tuberculosis (MDR-TB), is resistant to these first-line drugs. Here, two molecular techniques were demonstrated such as PCR sequencing-based and GeneXpert assay for rapidly identifying MDR-TB. Materials and Methods Pulmonary samples (sputum) were collected from 55 MDR-TB suspected patients from the National Tuberculosis Reference Laboratory (NTRL), Dhaka where the research work was partially accomplished and continued in the department of Microbiology, University of Dhaka, Bangladesh. We strived for sequencing technique as well as GeneXpert assay to identify mutations in rpoB and katG genes in MTB strains and sputum directly. Culture-based drug susceptibility testing (DST) was performed to measure the efficacy of the molecular methods employed. Results When analyzed, rpoB gene mutations at codons 531 (54.54%), 526 (14.54%), and 516 (10.91%) were found by sequencing in 80% of the samples. Nucleotide substitution at katG315 (AGC→ACC) was spotted in 16 (76.19%) out of 21 samples. When comparing the sequencing results with DST, sensitivity and specificity were investigated to determine drug-resistance (rifampicin-resistance were 98 and 100% whereas isoniazid-resistance were 94 and 100% respectively). Additionally, as a point of comparison with DST, only 85.45% of RIF mono-resistant TB cases were accurately evaluated by the GeneXpert assay. Conclusion This research supports the adoption of PCR sequencing approach as an efficient tool in detecting MDR-TB, counting the higher sensitivity and specificity as well as the short period to produce the results.
Collapse
Affiliation(s)
- Monika Sultana
- Department of Microbiology, University of Dhaka, Dhaka-1000, Bangladesh
| | | | | | - S M Mostafa Kamal
- National Tuberculosis Reference Laboratory (NTRL), Dhaka-1207, Bangladesh
| | | | - Mahmuda Yasmin
- Department of Microbiology, University of Dhaka, Dhaka-1000, Bangladesh
| |
Collapse
|
7
|
Harrison GA, Wang ER, Cho K, Mreyoud Y, Sarkar S, Almqvist F, Patti GJ, Stallings CL. Inducing vulnerability to InhA inhibition restores isoniazid susceptibility in drug-resistant Mycobacterium tuberculosis. mBio 2024; 15:e0296823. [PMID: 38294237 PMCID: PMC10936210 DOI: 10.1128/mbio.02968-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024] Open
Abstract
Of the approximately 10 million cases of Mycobacterium tuberculosis (Mtb) infections each year, over 10% are resistant to the frontline antibiotic isoniazid (INH). INH resistance is predominantly caused by mutations that decrease the activity of the bacterial enzyme KatG, which mediates the conversion of the pro-drug INH to its active form INH-NAD. We previously discovered an inhibitor of Mtb respiration, C10, that enhances the bactericidal activity of INH, prevents the emergence of INH-resistant mutants, and re-sensitizes a collection of INH-resistant mutants to INH through an unknown mechanism. To investigate the mechanism of action of C10, we exploited the toxicity of high concentrations of C10 to select for resistant mutants. We discovered two mutations that confer resistance to the disruption of energy metabolism and allow for the growth of Mtb in high C10 concentrations, indicating that growth inhibition by C10 is associated with inhibition of respiration. Using these mutants as well as direct inhibitors of the Mtb electron transport chain, we provide evidence that inhibition of energy metabolism by C10 is neither sufficient nor necessary to potentiate killing by INH. Instead, we find that C10 acts downstream of INH-NAD synthesis, causing Mtb to become particularly sensitive to inhibition of the INH-NAD target, InhA, without changing the concentration of INH-NAD or the activity of InhA, the two predominant mechanisms of potentiating INH. Our studies revealed that there exists a vulnerability in Mtb that can be exploited to render Mtb sensitive to otherwise subinhibitory concentrations of InhA inhibitor.IMPORTANCEIsoniazid (INH) is a critical frontline antibiotic to treat Mycobacterium tuberculosis (Mtb) infections. INH efficacy is limited by its suboptimal penetration of the Mtb-containing lesion and by the prevalence of clinical INH resistance. We previously discovered a compound, C10, that enhances the bactericidal activity of INH, prevents the emergence of INH-resistant mutants, and re-sensitizes a set of INH-resistant mutants to INH. Resistance is typically mediated by katG mutations that decrease the activation of INH, which is required for INH to inhibit the essential enzyme InhA. Our current work demonstrates that C10 re-sensitizes INH-resistant katG-hypomorphs without enhancing the activation of INH. We furthermore show that C10 causes Mtb to become particularly vulnerable to InhA inhibition without compromising InhA activity on its own. Therefore, C10 represents a novel strategy to curtail the development of INH resistance and to sensitize Mtb to sub-lethal doses of INH, such as those achieved at the infection site.
Collapse
Affiliation(s)
- Gregory A. Harrison
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Erin R. Wang
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yassin Mreyoud
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Souvik Sarkar
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Fredrik Almqvist
- Department of Chemistry, Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research, UCMR, Umeå University, Umeå, Sweden
| | - Gary J. Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Christina L. Stallings
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Uribe-Vázquez B, Díaz-Vilchis A, Avila-Linares A, Saab-Rincón G, Marín-Tovar Y, Flores H, Pastor N, Huerta-Miranda G, Rudiño-Piñera E, Soberón X. Characterization of a catalase-peroxidase variant (L333V-KatG) identified in an INH-resistant Mycobacterium tuberculosis clinical isolate. Biochem Biophys Rep 2024; 37:101649. [PMID: 38318524 PMCID: PMC10839757 DOI: 10.1016/j.bbrep.2024.101649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/24/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Mycobacterium tuberculosis catalase-peroxidase (Mt-KatG) is a bifunctional heme-dependent enzyme that has been shown to activate isoniazid (INH), the widely used antibiotic against tuberculosis (TB). The L333V-KatG variant has been associated with INH resistance in clinical M. tuberculosis isolates from Mexico. To understand better the mechanisms of INH activation, its catalytic properties (catalase, peroxidase, and IN-NAD formation) and crystal structure were compared with those of the wild-type enzyme (WT-KatG). The rate of IN-NAD formation mediated by WT-KatG was 23% greater than L333V-KatG when INH concentration is varied. In contrast to WT-KatG, the crystal structure of the L333V-KatG variant has a perhydroxy modification of the indole nitrogen of W107 from MYW adduct. L333V-KatG shows most of the active site residues in a similar position to WT-KatG; only R418 is in the R-conformation instead of the double R and Y conformation present in WT-KatG. L333V-KatG shows a small displacement respect to WT-KatG in the helix from R385 to L404 towards the mutation site, an increase in length of the coordination bond between H270 and heme Fe, and a longer H-bond between proximal D381 and W321, compared to WT-KatG; these small displacements could explain the altered redox potential of the heme, and result in a less active and stable enzyme.
Collapse
Affiliation(s)
- Brenda Uribe-Vázquez
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Adelaida Díaz-Vilchis
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Aylin Avila-Linares
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Gloria Saab-Rincón
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Yerli Marín-Tovar
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Humberto Flores
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Nina Pastor
- Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, UAEM, Avenida Universidad 1001, Colonia Chamilpa, 62209, Cuernavaca, Morelos, México
| | - Guillermo Huerta-Miranda
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Enrique Rudiño-Piñera
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| | - Xavier Soberón
- Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, UNAM, Avenida Universidad 2001, Colonia Chamilpa, 62210, Cuernavaca, México
| |
Collapse
|
9
|
Sampiron EG, Calsavara LL, Baldin VP, Montaholi DC, Leme ALD, Namba DY, Alves Olher VG, Caleffi-Ferraciolli KR, Cardoso RF, Siqueira VLD, Vandresen F, Scodro RBDL. Isoniazid-N-acylhydrazones as promising compounds for the anti-tuberculosis treatment. Tuberculosis (Edinb) 2023; 141:102363. [PMID: 37311289 DOI: 10.1016/j.tube.2023.102363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/29/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Tuberculosis (TB), a disease caused by Mycobacterium tuberculosis complex, still presents significant numbers of incidence and mortality, in addition to several cases of drug resistance. Resistance, especially to isoniazid, which is one of the main drugs used in the treatment, has increased. In this context, N-acylhydrazones derived from isoniazid have shown important anti-Mycobacterium tuberculosis activity. Hence, this work aimed to determine the anti-TB potential of 11 isoniazid-N-acylhydrazones (INH-acylhydrazones). For this purpose, the determination of minimum inhibitory concentration (MIC) against M. tuberculosis H37Rv and clinical isolates was carried out. Drug combination, minimum bactericidal concentration, cytotoxicity, and in silico parameters were also performed. INH-acylhydrazones (2), (8), and (9) had MIC for M. tuberculosis H37Rv similar to or lower than isoniazid, and bactericidal activity was observed. In addition, these compounds showed low cytotoxicity, with a selectivity index greater than 3,000. Interesting results were also obtained in the drug combination assay, with synergistic combinations with isoniazid, ethambutol, and rifampicin. In the in silico study, INH-acylhydrazones behaved similarly to INH, but with improvements in some aspects. Based on these findings, it is concluded that compounds (2), (8), and (9) are considered promising scaffolds and warrant further investigation for designing future antimicrobial drugs.
Collapse
Affiliation(s)
- Eloísa Gibin Sampiron
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, 87020-900, Brazil.
| | | | | | - Débora Cássia Montaholi
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, 87020-900, Brazil
| | | | - Danillo Yuji Namba
- Department of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86057-970, Brazil
| | | | - Katiany Rizzieri Caleffi-Ferraciolli
- Postgraduate Program in Bioscience and Physiopathology, UEM, Maringá, Paraná, 87020-900, Brazil; Department of Clinical Analysis and Biomedicine, UEM, Maringá, Paraná, 87020-900, Brazil
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, 87020-900, Brazil; Postgraduate Program in Bioscience and Physiopathology, UEM, Maringá, Paraná, 87020-900, Brazil; Department of Clinical Analysis and Biomedicine, UEM, Maringá, Paraná, 87020-900, Brazil
| | - Vera Lucia Dias Siqueira
- Postgraduate Program in Bioscience and Physiopathology, UEM, Maringá, Paraná, 87020-900, Brazil; Department of Clinical Analysis and Biomedicine, UEM, Maringá, Paraná, 87020-900, Brazil
| | - Fábio Vandresen
- Department of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86057-970, Brazil
| | - Regiane Bertin de Lima Scodro
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, 87020-900, Brazil; Department of Clinical Analysis and Biomedicine, UEM, Maringá, Paraná, 87020-900, Brazil
| |
Collapse
|
10
|
Abrahams KA, Batt SM, Gurcha SS, Veerapen N, Bashiri G, Besra GS. DprE2 is a molecular target of the anti-tubercular nitroimidazole compounds pretomanid and delamanid. Nat Commun 2023; 14:3828. [PMID: 37380634 DOI: 10.1038/s41467-023-39300-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/01/2023] [Indexed: 06/30/2023] Open
Abstract
Mycobacterium tuberculosis is one of the global leading causes of death due to a single infectious agent. Pretomanid and delamanid are new antitubercular agents that have progressed through the drug discovery pipeline. These compounds are bicyclic nitroimidazoles that act as pro-drugs, requiring activation by a mycobacterial enzyme; however, the precise mechanisms of action of the active metabolite(s) are unclear. Here, we identify a molecular target of activated pretomanid and delamanid: the DprE2 subunit of decaprenylphosphoribose-2'-epimerase, an enzyme required for the synthesis of cell wall arabinogalactan. We also provide evidence for an NAD-adduct as the active metabolite of pretomanid. Our results highlight DprE2 as a potential antimycobacterial target and provide a foundation for future exploration into the active metabolites and clinical development of pretomanid and delamanid.
Collapse
Affiliation(s)
- Katherine A Abrahams
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Sarah M Batt
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Sudagar S Gurcha
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Natacha Veerapen
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Ghader Bashiri
- Laboratory of Molecular and Microbial Biochemistry, School of Biological Sciences, University of Auckland, 3A Symonds Street, Auckland, 1010, New Zealand
| | - Gurdyal S Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
11
|
D'Elia JA, Weinrauch LA. Gated Calcium Ion Channel and Mutation Mechanisms in Multidrug-Resistant Tuberculosis. Int J Mol Sci 2023; 24:ijms24119670. [PMID: 37298620 DOI: 10.3390/ijms24119670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
A wide spectrum of Gram-positive/Gram-negative bacteria has been found resistant to a wide spectrum of antibiotics in the United States of America during the past decade. Drug-resistant tuberculosis is not yet a major threat in North/South America, Europe, and the Middle East. However, the migration of populations in times of drought, famine, and hostilities may increase the global reach of this ancient pathogen. Given an increased spread from China and India to African countries, drug-resistant Mycobacterium tuberculosis has become an emerging topic of concern for Europe and North America. Due to the dangers associated with the spread of pathogens among different populations, the World Health Organization continues to expand healthcare advisories for therapeutic approaches for both stationary and migrating populations. As much of the literature focuses on endemic to pandemic viruses, we remain concerned that other treatable communicable diseases may be ignored. One such disease is multidrug-resistant tuberculosis. We focus on molecular mechanisms that this pathogen relies upon for the development of multidrug resistance via gene mutation and the evolutionary development of new enzyme and calcium channels.
Collapse
Affiliation(s)
- John A D'Elia
- Kidney/Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Larry A Weinrauch
- Kidney/Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
12
|
Harrison GA, Cho K, Wang ER, Sarkar S, Almqvist F, Patti GJ, Stallings CL. Inducing vulnerability to InhA inhibition restores isoniazid susceptibility in drug resistant Mycobacterium tuberculosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527416. [PMID: 36798348 PMCID: PMC9934558 DOI: 10.1101/2023.02.06.527416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Of the approximately 10 million cases of Mycobacterium tuberculosis (Mtb) infections each year, over 10% are resistant to the frontline antibiotic isoniazid (INH). INH resistance is predominantly caused by mutations that decrease the activity of the bacterial enzyme KatG, which mediates conversion of the pro-drug INH to its active form INH-NAD. We previously discovered an inhibitor of Mtb respiration, C10, that enhances the bactericidal activity of INH, prevents the emergence of INH-resistant mutants, and re-sensitizes a collection of INH-resistant mutants to INH through an unknown mechanism. To investigate the mechanism of action of C10, we exploited the toxicity of high concentrations of C10 to select for resistant mutants. We discovered two mutations that confer resistance to the disruption of energy metabolism and allow for growth of Mtb in high C10 concentrations, indicating that growth inhibition by C10 is associated with inhibition of respiration. Using these mutants as well as direct inhibitors of the Mtb electron transport chain, we provide evidence that inhibition of energy metabolism by C10 is neither sufficient nor necessary to potentiate killing by INH. Instead, we find that C10 acts downstream of INH-NAD synthesis, causing Mtb to become particularly sensitive to inhibition of the INH-NAD target, InhA, without changing the concentration of INH-NAD or the activity of InhA, the two predominant mechanisms of potentiating INH. Our studies revealed that there exists a vulnerability in Mtb that can be exploited to render Mtb sensitive to otherwise subinhibitory concentrations of InhA inhibitor.
Collapse
Affiliation(s)
- Gregory A. Harrison
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO USA
| | - Erin R. Wang
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO, USA
| | - Souvik Sarkar
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Fredrik Almqvist
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden
- Umeå Centre for Microbial Research, UCMR, Umeå University, SE-90187 Umeå, Sweden
| | - Gary J. Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Center for Metabolomics and Isotope Tracing, Washington University in St. Louis, St. Louis, MO USA
| | - Christina L. Stallings
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
13
|
Kuppuswamy U, Rajan RK, Kumar A, Ramanathan M. In-silico and in-vitro analysis of novel substituted benzimidazolyl derivatives for antimycobacterial potentials targeting enoyl acyl carrier protein reductase (InhA). FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00449-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Background
The emergence of mutated drug-resistant strains of Mycobacterium tuberculosis has reinvigorated the development of effective chemotherapy for MDR-TB (multidrug-resistant resistance tuberculosis). Enoyl acyl carrier protein reductase (InhA) involved in the mycobacterial fatty acid elongation system has been chosen as a potential target.
Result
All of the lead compounds had a definite Rf value and a sharp melting point, confirming that no tautomeric forms exist and that the keto (CO) group is apparent in the IR and 13C NMR spectrum data. Structure-based drug design revealed the presence of amino acid residues like TYR 158, ILE 194, and PHE 149 which are crucial for InhA inhibitory activity and were considered favorable interactions. Among all, compounds 4, 5a, and 5c showed better docking and binding free energy owing to favorable interactions. Interestingly, there was a strong correlation between the binding free energy and the antimycobacterial susceptibility assay, where compounds 4, 5a, and 5c had greater activity. All the lead compounds also had good oral absorption and gut permeability. The presence of a carboxylic linker (–COOH–) between benzimidazole and the rest of the structure of the lead compounds was found to be crucial for activity as the oxygen atom and hydroxyl group of the linker formed most of the favorable interactions. The presence of chlorophenyl showed a favorable effect on InhA inhibition which might be owing to its hydrophobic interaction with PHE 149.
Conclusion
Three of the seven lead compounds synthesized had an IC value of approximately 0.5 μg/ml in the in-vitro Alamar blue assay against the Mycobacterium tuberculosis H37Rv strain, which is roughly comparable to the standard marketed drug, Isoniazid (INH). This manifestation of promising activity that resulted from combining in-silico and wet lab experimentation could be a great starting point for developing potent antimycobacterial agents to combat multidrug-resistant tuberculosis.
Graphical abstract
Collapse
|
14
|
Yan W, Zheng Y, Dou C, Zhang G, Arnaout T, Cheng W. The pathogenic mechanism of Mycobacterium tuberculosis: implication for new drug development. MOLECULAR BIOMEDICINE 2022; 3:48. [PMID: 36547804 PMCID: PMC9780415 DOI: 10.1186/s43556-022-00106-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a tenacious pathogen that has latently infected one third of the world's population. However, conventional TB treatment regimens are no longer sufficient to tackle the growing threat of drug resistance, stimulating the development of innovative anti-tuberculosis agents, with special emphasis on new protein targets. The Mtb genome encodes ~4000 predicted proteins, among which many enzymes participate in various cellular metabolisms. For example, more than 200 proteins are involved in fatty acid biosynthesis, which assists in the construction of the cell envelope, and is closely related to the pathogenesis and resistance of mycobacteria. Here we review several essential enzymes responsible for fatty acid and nucleotide biosynthesis, cellular metabolism of lipids or amino acids, energy utilization, and metal uptake. These include InhA, MmpL3, MmaA4, PcaA, CmaA1, CmaA2, isocitrate lyases (ICLs), pantothenate synthase (PS), Lysine-ε amino transferase (LAT), LeuD, IdeR, KatG, Rv1098c, and PyrG. In addition, we summarize the role of the transcriptional regulator PhoP which may regulate the expression of more than 110 genes, and the essential biosynthesis enzyme glutamine synthetase (GlnA1). All these enzymes are either validated drug targets or promising target candidates, with drugs targeting ICLs and LAT expected to solve the problem of persistent TB infection. To better understand how anti-tuberculosis drugs act on these proteins, their structures and the structure-based drug/inhibitor designs are discussed. Overall, this investigation should provide guidance and support for current and future pharmaceutical development efforts against mycobacterial pathogenesis.
Collapse
Affiliation(s)
- Weizhu Yan
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Yanhui Zheng
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Chao Dou
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Guixiang Zhang
- grid.13291.380000 0001 0807 1581Division of Gastrointestinal Surgery, Department of General Surgery and Gastric Cancer center, West China Hospital, Sichuan University, No. 37. Guo Xue Xiang, Chengdu, 610041 China
| | - Toufic Arnaout
- Kappa Crystals Ltd., Dublin, Ireland ,MSD Dunboyne BioNX, Co. Meath, Ireland
| | - Wei Cheng
- grid.412901.f0000 0004 1770 1022Division of Respiratory and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041 China
| |
Collapse
|
15
|
Kooti S, Abiri R, Kadivarian S, Khazayel S, Mohajeri P, Atashi S, Yari F, Ahmadpour H, Alvandi A. Modified gold nanoparticle colorimetric probe-based biosensor coupled with allele-specific PCR for rapid detection of G944C mutation associated with isoniazid resistance. SENSING AND BIO-SENSING RESEARCH 2022. [DOI: 10.1016/j.sbsr.2022.100543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
16
|
Abstract
Whole-genome sequencing (WGS) has shown tremendous potential in rapid diagnosis of drug-resistant tuberculosis (TB). In the current study, we performed WGS on drug-resistant Mycobacterium tuberculosis isolates obtained from Shanghai (n = 137) and Russia (n = 78). We aimed to characterise the underlying and high-frequency novel drug-resistance-conferring mutations, and also create valuable combinations of resistance mutations with high predictive sensitivity to predict multidrug- and extensively drug-resistant tuberculosis (MDR/XDR-TB) phenotype using a bootstrap method. Most strains belonged to L2.2, L4.2, L4.4, L4.5 and L4.8 lineages. We found that WGS could predict 82.07% of phenotypically drug-resistant domestic strains. The prediction sensitivity for rifampicin (RIF), isoniazid (INH), ethambutol (EMB), streptomycin (STR), ofloxacin (OFL), amikacin (AMK) and capreomycin (CAP) was 79.71%, 86.30%, 76.47%, 88.37%, 83.33%, 70.00% and 70.00%, respectively. The mutation combination with the highest sensitivity for MDR prediction was rpoB S450L + rpoB H445A/P + katG S315T + inhA I21T + inhA S94A, with a sensitivity of 92.17% (0.8615, 0.9646), and the mutation combination with highest sensitivity for XDR prediction was rpoB S450L + katG S315T + gyrA D94G + rrs A1401G, with a sensitivity of 92.86% (0.8158, 0.9796). The molecular information presented here will be of particular value for the rapid clinical detection of MDR- and XDR-TB isolates through laboratory diagnosis.
Collapse
|
17
|
The cleavage kinetics of hydrazide derivatives of isoniazid by HPLC-UV/DAD and its impact on activity against Mycobacterium tuberculosis. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1188:123080. [PMID: 34923301 DOI: 10.1016/j.jchromb.2021.123080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022]
Abstract
Isoniazid is a first-line drug for the treatment of tuberculosis, a bacterial disease caused by Mycobacterium tuberculosis. Its terminal amino group is highly reactive, leading to significant metabolic deactivation, drug interactions and hepatotoxicity. It is speculated that the activity of isoniazid derivatives is, in part, related to the cleavage of the protecting group. Therefore, this study aimed to evaluate the cleavage characteristics of previously developed isoniazid derivatives through kinetic studies by high-performance liquid chromatography with ultraviolet-diode array detectio to establish a comparison between the rates of the process and the respective activities against M. tuberculosis. Chromatographic separations were performed on an XDB C18 column coupled to an XDB C18 precolumn. The mobile phase consisted of ultrapure water and acetonitrile in gradient mode. The flow rate was 1.0 mL/min, the injection volume was 20 μL, and the detection wavelengths were 230 nm (derivatives and isatins) and 270 nm (isoniazid). Incubation of derivatives was carried out for 5 days in 10 mmol/L phosphate buffer solution (pH 3.0, 7.4, 8.0) or in fetal bovine serum at 37 °C. The incubation reduced the concentration of the derivatives and led to the formation of isoniazid in a first-order kinetic reaction. Isoniazid formation was logarithmically correlated with the minimum inhibitory concentration of the derivatives. The results showed that higher cleavage rates are associated with greater activities against M. tuberculosis, providing important information for the development of future generations of isoniazid derivatives and for screening drug candidates for the treatment of tuberculosis.
Collapse
|
18
|
Edwards DJ, Duchene S, Pope B, Holt KE. SNPPar: identifying convergent evolution and other homoplasies from microbial whole-genome alignments. Microb Genom 2021; 7:000694. [PMID: 34874243 PMCID: PMC8767352 DOI: 10.1099/mgen.0.000694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Homoplasic SNPs are considered important signatures of strong (positive) selective pressure, and hence of adaptive evolution for clinically relevant traits such as antibiotic resistance and virulence. Here we present a new tool, SNPPar, for efficient detection and analysis of homoplasic SNPs from large whole genome sequencing datasets (>1000 isolates and/or >100 000 SNPs). SNPPar takes as input an SNP alignment, tree and annotated reference genome, and uses a combination of simple monophyly tests and ancestral state reconstruction (ASR, via TreeTime) to assign mutation events to branches and identify homoplasies. Mutations are annotated at the level of codon and gene, to facilitate analysis of convergent evolution. Testing on simulated data (120 Mycobacterium tuberculosis alignments representing local and global samples) showed SNPPar can detect homoplasic SNPs with very high specificity (zero false-positives in all tests) and high sensitivity (zero false-negatives in 89 % of tests). SNPPar analysis of three empirically sampled datasets (Elizabethkingia anophelis, Burkholderia dolosa and M. tuberculosis) produced results that were in concordance with previous studies, in terms of both individual homoplasies and evidence of convergence at the codon and gene levels. SNPPar analysis of a simulated alignment of ~64 000 genome-wide SNPs from 2000 M. tuberculosis genomes took ~23 min and ~2.6 GB of RAM to generate complete annotated results on a laptop. This analysis required ASR be conducted for only 1.25 % of SNPs, and the ASR step took ~23 s and 0.4 GB of RAM. SNPPar automates the detection and annotation of homoplasic SNPs efficiently and accurately from large SNP alignments. As demonstrated by the examples included here, this information can be readily used to explore the role of homoplasy in parallel and/or convergent evolution at the level of nucleotide, codon and/or gene.
Collapse
Affiliation(s)
- David J. Edwards
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Sebastián Duchene
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria, Australia
| | - Bernard Pope
- Melbourne Bioinformatics, The University of Melbourne, 187 Grattan Street, Carlton, Victoria, Australia,Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, 305 Grattan Street, Melbourne, Victoria, Australia,Department of Medicine, Central Clinical School, Monash University, Clayton, Victoria, Australia,Department of Surgery (Royal Melbourne Hospital), Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Victoria, Australia
| | - Kathryn E. Holt
- Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia,Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK,*Correspondence: Kathryn E. Holt,
| |
Collapse
|
19
|
Whole-genome sequencing as a tool for studying the microevolution of drug-resistant serial Mycobacterium tuberculosis isolates. Tuberculosis (Edinb) 2021; 131:102137. [PMID: 34673379 DOI: 10.1016/j.tube.2021.102137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/14/2021] [Accepted: 10/03/2021] [Indexed: 11/21/2022]
Abstract
Treatment of drug-resistant tuberculosis requires extended use of more toxic and less effective drugs and may result in retreatment cases due to failure, abandonment or disease recurrence. It is therefore important to understand the evolutionary process of drug resistance in Mycobacterium tuberculosis. We here in describe the microevolution of drug resistance in serial isolates from six previously treated patients. Drug resistance was initially investigated through phenotypic methods, followed by genotypic approaches. The use of whole-genome sequencing allowed the identification of mutations in the katG, rpsL and rpoB genes associated with drug resistance, including the detection of rare mutations in katG and mixed populations of strains. Molecular docking simulation studies of the impact of observed mutations on isoniazid binding were also performed. Whole-genome sequencing detected 266 single nucleotide polymorphisms between two isolates obtained from one patient, suggesting a case of exogenous reinfection. In conclusion, sequencing technologies can detect rare mutations related to drug resistance, identify subpopulations of resistant strains, and identify diverse populations of strains due to exogenous reinfection, thus improving tuberculosis control by guiding early implementation of appropriate clinical and therapeutic interventions.
Collapse
|
20
|
Calsavara LL, Hegeto LA, Sampiron EG, Costacurta GF, Murase LS, Souza JV, de Almeida AL, de S Santos NC, Siqueira VL, de L Scodro RB, Cardoso RF, Caleffi-Ferracioli KR. Rescue of streptomycin activity by piperine in Mycobacterium tuberculosis. Future Microbiol 2021; 16:623-633. [PMID: 34098743 DOI: 10.2217/fmb-2020-0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate the modulatory effect of piperine (PIP) on streptomycin (SM) activity in Mycobacterium tuberculosis (Mtb). Materials & methods: SM and PIP minimum inhibitory concentration (MIC) and combinatory activity were determined in Mtb H37Rv and in susceptible and resistant clinical isolates. Ethidium bromide accumulation assay and relative quantification of efflux pumps genes (rv1258c, rv1218c and rv2942), after SM and SM+PIP combination exposure, were also performed. Results: PIP concentration of 25 μg/ml (1/4× MIC) was able to inhibit efflux pumps activity, to modulate SM activity in Mtb, and conducted changes in the relative quantification of efflux pumps genes. Conclusion: SM+PIP combination was able to rescue the SM-susceptible MIC values in SM-resistant Mtb.
Collapse
Affiliation(s)
- Leonora L Calsavara
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Laíse A Hegeto
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Eloisa G Sampiron
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Giovana F Costacurta
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Letícia S Murase
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - João Vp Souza
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Aryadne L de Almeida
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Nathally C de S Santos
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Vera Ld Siqueira
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Regiane B de L Scodro
- Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Rosilene F Cardoso
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Postgraduate Program in Health Sciences, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| | - Katiany R Caleffi-Ferracioli
- Postgraduate Program in Bioscience & Physiopathology, State University of Maringá (UEM), Maringá, Paraná, Brazil.,Department of Clinical Analysis & Biomedicine, Laboratory of Medical Bacteriology, State University of Maringa, Parana, Brazil
| |
Collapse
|
21
|
Zampieri D, Mamolo MG. Hybridization Approach to Drug Discovery Inhibiting Mycobacterium tuberculosis-An Overview. Curr Top Med Chem 2021; 21:777-788. [PMID: 32814528 DOI: 10.2174/1568026620666200819151342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 11/22/2022]
Abstract
Tuberculosis is one of the top 10 causes of death worldwide and the leading cause of death from a single infectious agent, mainly due to Mycobacterium tuberculosis (MTB). Recently, clinical prognoses have worsened due to the emergence of multi-drug resistant (MDR) and extensive-drug resistant (XDR) tuberculosis, which lead to the need for new, efficient and safe drugs. Among the several strategies, polypharmacology could be considered one of the best solutions, in particular, the multitarget directed ligands strategy (MTDLs), based on the synthesis of hybrid ligands acting against two targets of the pathogen. The framework strategy comprises linking, fusing and merging approaches to develop new chemical entities. With these premises, this review aims to provide an overview of the recent hybridization approach, in medicinal chemistry, of the most recent and promising multitargeting antimycobacterial candidates.
Collapse
Affiliation(s)
- Daniele Zampieri
- Department of Chemical and Pharmaceutical Sciences, P.le Europa 1, University of Trieste, Trieste 34127, Italy
| | - Maria G Mamolo
- Department of Chemical and Pharmaceutical Sciences, P.le Europa 1, University of Trieste, Trieste 34127, Italy
| |
Collapse
|
22
|
Trinh TND, Lee NY. Nucleic acid amplification-based microfluidic approaches for antimicrobial susceptibility testing. Analyst 2021; 146:3101-3113. [PMID: 33876805 DOI: 10.1039/d1an00180a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Because of the global spread of antimicrobials, there is an urgent need to develop rapid and effective tools for antimicrobial susceptibility testing to help clinicians prescribe accurate and appropriate antibiotic doses sooner. The conventional methods for antimicrobial susceptibility testing are usually based on bacterial culture methods, which are time-consuming, complicated, and labor-intensive. Therefore, other approaches are needed to address these issues. Recently, microfluidic technology has gained significant attention in infection management due to its advantages including rapid detection, high sensitivity and specificity, highly automated assay, simplicity, low cost, and potential for point-of-care testing in low-resource areas. Microfluidic advances for antimicrobial susceptibility testing can be classified into phenotypic (usually culture-based) and genotypic tests. Genotypic antimicrobial susceptibility testing is the detection of resistant genes in a microorganism using methods such as nucleic acid amplification. This review (with 107 references) surveys the different forms of nucleic acid amplification-based microdevices used for genotypic antimicrobial susceptibility testing. The first section reviews the serious threat of antimicrobial-resistant microorganisms and the urgent need for fast check-ups. Next, several conventional antimicrobial susceptibility testing methods are discussed, and microfluidic technology as a promising candidate for rapid detection of antimicrobial-resistant microorganisms is briefly introduced. The next section highlights several advancements of microdevices, with an emphasis on their working principles and performance. The review concludes with the importance of fully integrated microdevices and a discussion on future perspectives.
Collapse
Affiliation(s)
- Thi Ngoc Diep Trinh
- Department of Industrial Environmental Engineering, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, Korea
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, Gyeonggi-do 13120, Korea.
| |
Collapse
|
23
|
Munir A, Wilson MT, Hardwick SW, Chirgadze DY, Worrall JAR, Blundell TL, Chaplin AK. Using cryo-EM to understand antimycobacterial resistance in the catalase-peroxidase (KatG) from Mycobacterium tuberculosis. Structure 2021; 29:899-912.e4. [PMID: 33444527 PMCID: PMC8355310 DOI: 10.1016/j.str.2020.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022]
Abstract
Resolution advances in cryoelectron microscopy (cryo-EM) now offer the possibility to visualize structural effects of naturally occurring resistance mutations in proteins and also of understanding the binding mechanisms of small drug molecules. In Mycobacterium tuberculosis the multifunctional heme enzyme KatG is indispensable for activation of isoniazid (INH), a first-line pro-drug for treatment of tuberculosis. We present a cryo-EM methodology for structural and functional characterization of KatG and INH resistance variants. The cryo-EM structure of the 161 kDa KatG dimer in the presence of INH is reported to 2.7 Å resolution allowing the observation of potential INH binding sites. In addition, cryo-EM structures of two INH resistance variants, identified from clinical isolates, W107R and T275P, are reported. In combination with electronic absorbance spectroscopy our cryo-EM approach reveals how these resistance variants cause disorder in the heme environment preventing heme uptake and retention, providing insight into INH resistance. A cryo-EM structure to 2.7 Å resolution of M. tuberculosis KatG with isoniazid Cryo-EM is able to visualize multiple dynamic binding modes of isoniazid to KatG Structural disorder in isoniazid resistance mutations is observed Structural disorder of the resistance mutations results in the lack of heme retention
Collapse
Affiliation(s)
- Asma Munir
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Michael T Wilson
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Steven W Hardwick
- CryoEM Facility, Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Dimitri Y Chirgadze
- CryoEM Facility, Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK
| | - Jonathan A R Worrall
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK.
| | - Amanda K Chaplin
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1GA, UK.
| |
Collapse
|
24
|
Butler TE, Lee AJ, Yang Y, Newton MD, Kargupta R, Puttaswamy S, Sengupta S. Direct-from-sputum rapid phenotypic drug susceptibility test for mycobacteria. PLoS One 2020; 15:e0238298. [PMID: 32857802 PMCID: PMC7454970 DOI: 10.1371/journal.pone.0238298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
Background The spread of multi-drug resistant tuberculosis (MDR-TB) is a leading global public-health challenge. Because not all biological mechanisms of resistance are known, culture-based (phenotypic) drug-susceptibility testing (DST) provides important information that influences clinical decision-making. Current phenotypic tests typically require pre-culture to ensure bacterial loads are at a testable level (taking 2–4 weeks) followed by 10–14 days to confirm growth or lack thereof. Methods and findings We present a 2-step method to obtain DST results within 3 days of sample collection. The first involves selectively concentrating live mycobacterial cells present in relatively large volumes of sputum (~2-10mL) using commercially available magnetic-nanoparticles (MNPs) into smaller volumes, thereby bypassing the need for pre-culture. The second involves using microchannel Electrical Impedance Spectroscopy (m-EIS) to monitor multiple aliquots of small volumes (~10μL) of suspension containing mycobacterial cells, MNPs, and candidate-drugs to determine whether cells grow, die, or remain static under the conditions tested. m-EIS yields an estimate for the solution “bulk capacitance” (Cb), a parameter that is proportional to the number of live bacteria in suspension. We are thus able to detect cell death (bactericidal action of the drug) in addition to cell-growth. We demonstrate proof-of-principle using M. bovis BCG and M. smegmatis suspended in artificial sputum. Loads of ~ 2000–10,000 CFU of mycobacteria were extracted from ~5mL of artificial sputum during the decontamination process with efficiencies of 84% -100%. Subsequently, suspensions containing ~105 CFU/mL of mycobacteria with 10 mg/mL of MNPs were monitored in the presence of bacteriostatic and bactericidal drugs at concentrations below, at, and above known MIC (Minimum Inhibitory Concentration) values. m-EIS data (ΔCb) showed data consistent with growth, death or stasis as expected and/or recorded using plate counts. Electrical signals of death were visible as early as 3 hours, and growth was seen in < 3 days for all samples, allowing us to perform DST in < 3 days. Conclusion We demonstrated “proof of principle” that (a) live mycobacteria can be isolated from sputum using MNPs with high efficiency (almost all the bacteria that survive decontamination) and (b) that the efficacy of candidate drugs on the mycobacteria thus isolated (in suspensions containing MNPs) could be tested in real-time using m-EIS.
Collapse
Affiliation(s)
- Timothy E. Butler
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Aiden J. Lee
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Yongqiang Yang
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
| | | | - Roli Kargupta
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Sachidevi Puttaswamy
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
| | - Shramik Sengupta
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
25
|
Veeravarapu H, Malkhed V, Mustyala KK, Vadija R, Malikanti R, Vuruputuri U, Muthyala MKK. Structure-based drug design, synthesis and screening of MmaA1 inhibitors as novel anti-TB agents. Mol Divers 2020; 25:351-366. [PMID: 32533514 DOI: 10.1007/s11030-020-10107-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/15/2020] [Indexed: 02/05/2023]
Abstract
Tuberculosis is one of the leading causes of death across the world. The treatment regimens for tuberculosis are well established, but still the control of the disease faces many challenges such as lengthy treatment protocols, drug resistance and toxicity. In the present work, mycolic acid methyl transferase (MmaA1), a protein involved in the maturation of mycolic acids in the biochemical pathway of the Mycobacterium, was studied for novel drug discovery. The homology model of the MmaA1 protein was built and validated by using computational techniques. The MmaA1 protein has 286 amino acid residues consisting of 10 α-helices and 7 β-sheets. The active site of the MmaA1 protein was identified using CASTp, SiteMap and PatchDock. Virtual screening studies were performed with two small molecule ligand databases: Asinex synergy and Diverse_Elite_Gold_Platinum databases having a total of 43,446 molecules and generated 1,30,814 conformers against the predicted and validated active site of the MmaA1 protein. Binding analysis showed that the residues ASP 19, PHE 22, TRP 30, TYR 32, TRP 74 and ALA 77 of MmaA1 protein have consistent interactions with the ligands. The hit ligands were further filtered by in silico ADME properties to eliminate potentially toxic molecules. Of the top 10 molecules, 3-(2-morpholinoacetamido)-N-(1,4-dihydro-4-oxoquinazolin-6-yl) benzamide was synthesised and screened for in vitro anti-TB activity against Mtb H37Rv using MABA assay. The compound and its intermediates exhibited good in vitro anti-TB activity which can be taken up for future lead optimisation studies. Structure based virtual screening study was performed using a validated homology model against small molecules from two virtual compound libraries. Synthesised the lead compound 3-(2-morpholinoacetamido)-N-(1,4-dihydro-4-oxoquinazolin-6-yl)benzamide obtained from virtual screening. In vitro activity against Mtb H37Rv has given a promising result.
Collapse
Affiliation(s)
- Hymavathi Veeravarapu
- Pharmaceutical Chemistry Research Lab, AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India.,Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Vasavi Malkhed
- Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Kiran Kumar Mustyala
- Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Rajender Vadija
- Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Ramesh Malikanti
- Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Uma Vuruputuri
- Molecular Modelling Research Lab, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, India
| | - Murali Krishna Kumar Muthyala
- Pharmaceutical Chemistry Research Lab, AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, 530003, India.
| |
Collapse
|
26
|
AlMatar M, Var I, Kayar B, Köksal F. Differential Expression of Resistant and Efflux Pump Genes in MDR-TB Isolates. Endocr Metab Immune Disord Drug Targets 2020; 20:271-287. [DOI: 10.2174/1871530319666191009153834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/21/2019] [Accepted: 07/22/2019] [Indexed: 12/20/2022]
Abstract
Background:Numerous investigations demonstrate efflux as a worldwide bacterial mode of action which contributes to the resistance of drugs. The activity of antibiotics, which subjects to efflux, can be improved by the combined usage of efflux inhibitors. However, the efflux role to the overall levels of antibiotic resistance of clinical M. tuberculosis isolates is inadequately comprehended and is still disregarded by many.Method:Here, we assessed the contribution of resistant genes associated with isoniazid (INH) and rifampin (R) resistance to the levels of drug resistance in the (27) clinical isolates of MDR-TB. Additionally, the role of the resistance for six putative drug efflux pump genes to the antibiotics was investigated. The level of katG expression was down-regulated in 24/27 (88.88%) of MDR-TB isolates. Of the 27 MDR-TB isolates, inhA, oxyR-ahpC, and rpoB showed either overexpression or up-regulation in 8 (29.62%), 4 (14.81 %), and 24 (88.88%), respectively. Moreover, the efflux pump genes drrA, drrB, efpA, Rv2459, Rv1634, and Rv1250 were overexpressed under INH/RIF plus fresh pomegranate juice (FPJ) stress signifying the efflux pumps contribution to the overall levels of the resistance of MDR-TB isolates.Conclusion:These results displayed that the levels of drug resistance of MDR-TB clinical isolates are due to combination among drug efflux pump and the presence of mutations in target genes, a truth which is often ignored by the specialists of tuberculosis in favour of the almost undoubted significance of drug target- gene mutations for the resistance in M. tuberculosis.
Collapse
Affiliation(s)
- Manaf AlMatar
- Department of Biotechnology, Institute of Natural and Applied Sciences (Fen Bilimleri Enstitusu), Cukurova University, Adana, Turkey
| | - Işıl Var
- Department of Food Engineering, Agricultural Faculty, Cukurova University, Adana, Turkey
| | - Begüm Kayar
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Fatih Köksal
- Department of Medical Microbiology, Faculty of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
27
|
Differential Sensitivity of Mycobacteria to Isoniazid Is Related to Differences in KatG-Mediated Enzymatic Activation of the Drug. Antimicrob Agents Chemother 2020; 64:AAC.01899-19. [PMID: 31767723 DOI: 10.1128/aac.01899-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/19/2019] [Indexed: 11/20/2022] Open
Abstract
Isoniazid (INH) is a cornerstone of antitubercular therapy. Mycobacterium tuberculosis complex bacteria are the only mycobacteria sensitive to clinically relevant concentrations of INH. All other mycobacteria, including M. marinum and M. avium subsp. paratuberculosis are resistant. INH requires activation by bacterial KatG to inhibit mycobacterial growth. We tested the role of the differences between M. tuberculosis KatG and that of other mycobacteria in INH sensitivity. We cloned the M. bovis katG gene into M. marinum and M. avium subsp. paratuberculosis and measured the MIC of INH. We recombinantly expressed KatG of these mycobacteria and tested in vitro binding to, and activation of, INH. Introduction of katG from M. bovis into M. marinum and M. avium subsp. paratuberculosis rendered them 20 to 30 times more sensitive to INH. Analysis of different katG sequences across the genus found KatG evolution diverged from RNA polymerase-defined mycobacterial evolution. Biophysical and biochemical tests of M. bovis and nontuberculous mycobacteria (NTM) KatG proteins showed lower affinity to INH and substantially lower enzymatic capacity for the conversion of INH into the active form in NTM. The KatG proteins of M. marinum and M. avium subsp. paratuberculosis are substantially less effective in INH activation than that of M. tuberculosis, explaining the relative INH insensitivity of these microbes. These data indicate that the M. tuberculosis complex KatG is divergent from the KatG of NTM, with a reciprocal relationship between resistance to host defenses and INH resistance. Studies of bacteria where KatG is functionally active but does not activate INH may aid in understanding M. tuberculosis INH-resistance mechanisms, and suggest paths to overcome them.
Collapse
|
28
|
Pharmacoinformatics-based identification of anti-bacterial catalase-peroxidase enzyme inhibitors. Comput Biol Chem 2019; 83:107136. [DOI: 10.1016/j.compbiolchem.2019.107136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/28/2019] [Accepted: 09/29/2019] [Indexed: 11/17/2022]
|
29
|
Johnson LA, Hug LA. Distribution of reactive oxygen species defense mechanisms across domain bacteria. Free Radic Biol Med 2019; 140:93-102. [PMID: 30930298 DOI: 10.1016/j.freeradbiomed.2019.03.032] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/06/2019] [Accepted: 03/26/2019] [Indexed: 11/25/2022]
Abstract
Bacteria are the most diverse and numerous organisms on the planet, inhabiting environments from the deep subsurface to particles in clouds. Across this range of conditions, bacteria have evolved a diverse suite of enzymes to mitigate cellular damage from reactive oxygen species (ROS). Here, we review the diversity and distribution of ROS enzymatic defense mechanisms across the domain Bacteria, using both peer-reviewed literature and publicly available genome databases. We describe the specific strategies used by well-characterized organisms in order to highlight differences in oxidative stress responses between aerobic, facultatively anaerobic, and anaerobic lifestyles. We present evidence from genome minimization experiments to suggest that ROS defenses are obligately required for life. This review clarifies the variability in ROS defenses across Bacteria, including the novel diversity found in currently uncharacterized Candidate Phyla.
Collapse
Affiliation(s)
- Lisa A Johnson
- Department of Biology, University of Waterloo, Waterloo, Canada
| | - Laura A Hug
- Department of Biology, University of Waterloo, Waterloo, Canada.
| |
Collapse
|
30
|
Pal R, Hameed S, Kumar P, Singh S, Fatima Z. Understanding lipidomic basis of iron limitation induced chemosensitization of drug-resistant Mycobacterium tuberculosis. 3 Biotech 2019; 9:122. [PMID: 30863701 PMCID: PMC6401079 DOI: 10.1007/s13205-019-1645-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Under limited micronutrients condition, Mycobacterium tuberculosis (MTB) has to struggle for acquisition of the limited micronutrients available in the host. One such crucial micronutrient that MTB requires for the growth and sustenance is iron. The present study aimed to sequester the iron supply of MTB to control drug resistance in MTB. We found that iron restriction renders hypersensitivity to multidrug-resistant MTB strains against first-line anti-TB drugs. To decipher the effect of iron restriction on possible mechanisms of chemosensitization and altered cellular circuitry governing drug resistance and virulence of MTB, we explored MTB cellular architecture. We could identify non-intact cell envelope, tampered MTB morphology and diminished mycolic acid under iron restricted MDR-MTB cells. Deeper exploration unraveled altered lipidome profile observed through conventional TLC and advanced mass spectrometry-based LC-ESI-MS techniques. Lipidome analysis not only depicted profound alterations of various lipid classes which are crucial for pathogenecity but also exposed leads such as indispensability of iron to sustain metabolic, genotoxic and oxidative stresses. Furthermore, iron deprivation led to inhibited biofilm formation and capacity of MTB to adhere buccal epithelial cells. Lastly, we demonstrated enhanced survival of Mycobacterium-infected Caenorhabditis elegans model under iron limitation. The present study offers evidence and proposes alteration of lipidome profile and affected virulence traits upon iron chelation. Taken together, iron deprivation could be a potential strategy to rescue MDR and enhance the effectiveness of existing anti-TB drugs.
Collapse
Affiliation(s)
- Rahul Pal
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| | - Saif Hameed
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| | - Parveen Kumar
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Sarman Singh
- Division of Clinical Microbiology and Molecular Medicine, Department of Laboratory Medicine, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Zeeshan Fatima
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram, 122413 India
| |
Collapse
|
31
|
Clerc DG. Extending the drug discovery pipeline to simultaneously-applied chemical agents, and extending the study of evolution to simultaneous mutations, through an ab initio model that relates changes in phenotype to changes in molecular binding interactions. INFORMATICS IN MEDICINE UNLOCKED 2019. [DOI: 10.1016/j.imu.2019.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
32
|
Validation of Novel Mycobacterium tuberculosis Isoniazid Resistance Mutations Not Detectable by Common Molecular Tests. Antimicrob Agents Chemother 2018; 62:AAC.00974-18. [PMID: 30082293 DOI: 10.1128/aac.00974-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/03/2018] [Indexed: 01/20/2023] Open
Abstract
Resistance to the first-line antituberculosis (TB) drug isoniazid (INH) is widespread, and the mechanism of resistance is unknown in approximately 15% of INH-resistant (INH-R) strains. To improve molecular detection of INH-R TB, we used whole-genome sequencing (WGS) to analyze 52 phenotypically INH-R Mycobacterium tuberculosis complex (MTBC) clinical isolates that lacked the common katG S315T or inhA promoter mutations. Approximately 94% (49/52) of strains had mutations at known INH-associated loci that were likely to confer INH resistance. All such mutations would be detectable by sequencing more DNA adjacent to existing target regions. Use of WGS minimized the chances of missing infrequent INH resistance mutations outside commonly targeted hotspots. We used recombineering to generate 12 observed clinical katG mutations in the pansusceptible H37Rv reference strain and determined their impact on INH resistance. Our functional genetic experiments have confirmed the role of seven suspected INH resistance mutations and discovered five novel INH resistance mutations. All recombineered katG mutations conferred resistance to INH at a MIC of ≥0.25 μg/ml and should be added to the list of INH resistance determinants targeted by molecular diagnostic assays. We conclude that WGS is a useful tool for detecting uncommon INH resistance mutations that would otherwise be missed by current targeted molecular testing methods and suggest that its use (or use of expanded conventional or next-generation-based targeted sequencing) may provide earlier diagnosis of INH-R TB.
Collapse
|
33
|
Sarkar R, Mdladla C, Macingwana L, Pietersen RD, Ngwane A, Tabb D, van Helden P, Wiid I, Baker B. Proteomic analysis reveals that sulfamethoxazole induces oxidative stress in M. tuberculosis. Tuberculosis (Edinb) 2018; 111:78-85. [DOI: 10.1016/j.tube.2018.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/27/2018] [Accepted: 05/15/2018] [Indexed: 02/04/2023]
|
34
|
Kumwenda GP, Chipungu G, Sloan DJ, Kaimila Y, Chiumya K, Pangani H. The occurrence and frequency of genomic mutations that mediate Isoniazid and Rifampicin resistance in Mycobacterium tuberculosis isolates from untreated pulmonary Tuberculosis cases in urban Blantyre, Malawi. Malawi Med J 2018; 30:1-5. [PMID: 29868151 PMCID: PMC5974378 DOI: 10.4314/mmj.v30i1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background The emergence and spread of drug-resistant Tuberculosis (TB) is a major public health threat. TB resistance originates in the course of treatment due to genomic mutations in Mycobacterium tuberculosis (MTB). An increase in new cases with drug-resistant TB could be an indicator of high levels of circulating resistant strains. This study was conducted to determine the occurrence and frequency of genomic mutations that mediate Isoniazid (INH) and Rifampicin (RIF) resistance among isolates from untreated TB cases in urban Blantyre, Malawi. Methods A cross-sectional retrospective study was conducted on a panel of 141(n=141) MTB clinical isolates recovered between June 2010 and January 2012 from >2+ Ziehl-Neelsen smear positive new pulmonary-TB patients with no history of treatment. Frozen isolates were revived using the BACTEC MGIT detection system. DNA was extracted using GenoLyse DNA extraction kit and detection of genomic mutations was carried out using the GenoType MTBDRplus Ver 2.0 assay. Results Out of the 141 isolates studied, 3 (2.1%) were found carrying mutations in the katG gene that confer resistance to Isoniazid (INH). No mutations were detected in the inhA promoter region gene that confer weak INH resistance or in the rpoB gene that confer Rifampicin resistance. All katG mutant genes had a S315T1 single point mutation, a genomic alteration that mediates high INH resistance. Conclusion The katG mutant gene conferring resistance to INH was the only genomic mutation observed among the isolates studied and the frequency of occurrence was low. Our findings suggest low levels of circulating drug-resistant MTB strains in urban Blantyre, Malawi.
Collapse
Affiliation(s)
| | | | | | - Yankho Kaimila
- The University of Malawi, College of Medicine, Blantyre, Malawi
| | - Kondwani Chiumya
- The University of Malawi, College of Medicine - Wellcome Trust TB Research Laboratory, Blantyre, Malawi
| | - Harry Pangani
- The University of Malawi, College of Medicine - Wellcome Trust TB Research Laboratory, Blantyre, Malawi
| |
Collapse
|
35
|
Nieto R LM, Mehaffy C, Islam MN, Fitzgerald B, Belisle J, Prenni J, Dobos K. Biochemical Characterization of Isoniazid-resistant Mycobacterium tuberculosis: Can the Analysis of Clonal Strains Reveal Novel Targetable Pathways? Mol Cell Proteomics 2018; 17:1685-1701. [PMID: 29844232 DOI: 10.1074/mcp.ra118.000821] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Indexed: 01/01/2023] Open
Abstract
Tuberculosis (TB) continues to be an important public health threat worldwide, due in part to drug resistant Mycobacterium tuberculosis (Mtb) strains. The United States recently reported a shortage of isoniazid (INH), which could drive higher INH resistance rates. Changes in the Mtb proteome before and after acquisition of INH resistance in a clean genetic background remain understudied and may elucidate alternate drug targets. Here, we focused on Mtb clonal strains to characterize the consequences of INH resistance on mycobacterial metabolism. Proteomic analysis was conducted by liquid-chromatography tandem mass spectrometry (LC-MS/MS) of cellular and secreted fractions, followed by a normalized spectral counting (NSAF) analysis (data are available via ProteomeXchange with identifier PXD009549). Two different Mtb clonal pairs representing a specific genetic lineage (one clinical and one generated in the laboratory) but sharing a katG mutation associated with INH resistance, were used in our analysis. Overall, we found 26 Mtb proteins with altered abundances after acquisition of INH resistance across both Mtb genetic lineages studied. These proteins were involved in ATP synthesis, lipid metabolism, regulatory events, and virulence, detoxification, and adaptation processes. Proteomic findings were validated by Western blotting analyses whenever possible. Mycolic acid (MA) analysis through LC/MS in the clonal Mtb pairs did not reveal a common trend in the alteration of these fatty acids across both INHr strains but revealed a significant reduction in levels of the two more abundant α-MA features in the clinical INHr strain. Interestingly, the clinical clonal pair demonstrated more variation in the abundance of the proteins involved in the FAS II pathway. Together, the proteomic and lipidomic data highlight the identification of potential drug targets such as alternative lipid biosynthetic pathways that may be exploited to combat clinically relevant Mtb INHr strains.
Collapse
Affiliation(s)
| | | | - M Nurul Islam
- From the ‡Department of Microbiology, Immunology and Pathology
| | | | - John Belisle
- From the ‡Department of Microbiology, Immunology and Pathology
| | - Jessica Prenni
- §Proteomics and Metabolomics Facility, Colorado State University, Fort Collins, CO
| | - Karen Dobos
- From the ‡Department of Microbiology, Immunology and Pathology,
| |
Collapse
|
36
|
Pienaar E, Linderman JJ, Kirschner DE. Emergence and selection of isoniazid and rifampin resistance in tuberculosis granulomas. PLoS One 2018; 13:e0196322. [PMID: 29746491 PMCID: PMC5944939 DOI: 10.1371/journal.pone.0196322] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/11/2018] [Indexed: 12/15/2022] Open
Abstract
Drug resistant tuberculosis is increasing world-wide. Resistance against isoniazid (INH), rifampicin (RIF), or both (multi-drug resistant TB, MDR-TB) is of particular concern, since INH and RIF form part of the standard regimen for TB disease. While it is known that suboptimal treatment can lead to resistance, it remains unclear how host immune responses and antibiotic dynamics within granulomas (sites of infection) affect emergence and selection of drug-resistant bacteria. We take a systems pharmacology approach to explore resistance dynamics within granulomas. We integrate spatio-temporal host immunity, INH and RIF dynamics, and bacterial dynamics (including fitness costs and compensatory mutations) in a computational framework. We simulate resistance emergence in the absence of treatment, as well as resistance selection during INH and/or RIF treatment. There are four main findings. First, in the absence of treatment, the percentage of granulomas containing resistant bacteria mirrors the non-monotonic bacterial dynamics within granulomas. Second, drug-resistant bacteria are less frequently found in non-replicating states in caseum, compared to drug-sensitive bacteria. Third, due to a steeper dose response curve and faster plasma clearance of INH compared to RIF, INH-resistant bacteria have a stronger influence on treatment outcomes than RIF-resistant bacteria. Finally, under combination therapy with INH and RIF, few MDR bacteria are able to significantly affect treatment outcomes. Overall, our approach allows drug-specific prediction of drug resistance emergence and selection in the complex granuloma context. Since our predictions are based on pre-clinical data, our approach can be implemented relatively early in the treatment development process, thereby enabling pro-active rather than reactive responses to emerging drug resistance for new drugs. Furthermore, this quantitative and drug-specific approach can help identify drug-specific properties that influence resistance and use this information to design treatment regimens that minimize resistance selection and expand the useful life-span of new antibiotics.
Collapse
Affiliation(s)
- Elsje Pienaar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer J. Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
37
|
Abbadi BL, Rodrigues-Junior VDS, Dadda ADS, Pissinate K, Villela AD, Campos MM, Lopes LGDF, Bizarro CV, Machado P, Sousa EHS, Basso LA. Is IQG-607 a Potential Metallodrug or Metallopro-Drug With a Defined Molecular Target in Mycobacterium tuberculosis? Front Microbiol 2018; 9:880. [PMID: 29765372 PMCID: PMC5938375 DOI: 10.3389/fmicb.2018.00880] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022] Open
Abstract
The emergence of strains of Mycobacterium tuberculosis resistant to isoniazid (INH) has underscored the need for the development of new anti-tuberculosis agents. INH is activated by the mycobacterial katG-encoded catalase-peroxidase, forming an acylpyridine fragment that is covalently attached to the C4 of NADH. This isonicotinyl-NAD adduct inhibits the activity of 2-trans-enoyl-ACP(CoA) reductase (InhA), which plays a role in mycolic acid biosynthesis. A metal-based INH analog, Na3[FeII(CN)5(INH)]·4H2O, IQG-607, was designed to have an electronic redistribution on INH moiety that would lead to an intramolecular electron transfer to bypass KatG activation. HPLC and EPR studies showed that the INH moiety can be oxidized by superoxide or peroxide yielding similar metabolites and isonicotinoyl radical only when associated to IQG-607, thereby supporting redox-mediated drug activation as a possible mechanism of action. However, IQG-607 was shown to inhibit the in vitro activity of both wild-type and INH-resistant mutant InhA enzymes in the absence of KatG activation. IQG-607 given by the oral route to M. tuberculosis-infected mice reduced lung lesions. Experiments using early and late controls of infection revealed a bactericidal activity for IQG-607. HPLC and voltammetric methods were developed to quantify IQG-607. Pharmacokinetic studies showed short half-life, high clearance, moderate volume of distribution, and low oral bioavailability, which was not altered by feeding. Safety and toxic effects of IQG-607 after acute and 90-day repeated oral administrations in both rats and minipigs showed occurrence of mild to moderate toxic events. Eight multidrug-resistant strains (MDR-TB) were resistant to IQG-607, suggesting an association between katG mutation and increasing MIC values. Whole genome sequencing of three spontaneous IQG-607-resistant strains harbored katG gene mutations. MIC measurements and macrophage infection experiments with a laboratorial strain showed that katG mutation is sufficient to confer resistance to IQG-607 and that the macrophage intracellular environment cannot trigger the self-activation mechanism. Reduced activity of IQG-607 against an M. tuberculosis strain overexpressing S94A InhA mutant protein suggested both the need for KatG activation and InhA as its target. Further efforts are suggested to be pursued toward attempting to translate IQG-607 into a chemotherapeutic agent to treat tuberculosis.
Collapse
Affiliation(s)
- Bruno L Abbadi
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Valnês da Silva Rodrigues-Junior
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adilio da Silva Dadda
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Kenia Pissinate
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Anne D Villela
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Maria M Campos
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz G de França Lopes
- Grupo de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Cristiano V Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pablo Machado
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo H S Sousa
- Grupo de Bioinorgânica, Departamento de Química Orgânica e Inorgânica, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Luiz A Basso
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Medicina e Ciências da Saúde, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
38
|
Loewen P, De Silva PM, Donald LJ, Switala J, Villanueva J, Fita I, Kumar A. KatG-Mediated Oxidation Leading to Reduced Susceptibility of Bacteria to Kanamycin. ACS OMEGA 2018; 3:4213-4219. [PMID: 29732452 PMCID: PMC5928485 DOI: 10.1021/acsomega.8b00356] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Resistance to antibiotics has become a serious problem for society, and there are increasing efforts to understand the reasons for and sources of resistance. Bacterial-encoded enzymes and transport systems, both innate and acquired, are the most frequent culprits for the development of resistance, although in Mycobacterium tuberculosis, the catalase-peroxidase, KatG, has been linked to the activation of the antitubercular drug isoniazid. While investigating a possible link between aminoglycoside antibiotics and the induction of oxidative bursts, we observed that KatG reduces susceptibility to aminoglycosides. Investigation revealed that kanamycin served as an electron donor for the peroxidase reaction, reducing the oxidized ferryl intermediates of KatG to the resting state. Loss of electrons from kanamycin was accompanied by the addition of a single oxygen atom to the aminoglycoside. The oxidized form of kanamycin proved to be less effective as an antibiotic. Kanamycin inhibited the crystallization of KatG, but the smaller, structurally related glycoside maltose did cocrystallize with KatG, providing a suggestion as to the possible binding site of kanamycin.
Collapse
Affiliation(s)
- Peter
C. Loewen
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - P. Malaka De Silva
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - Lynda J. Donald
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - Jacek Switala
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - Jacylyn Villanueva
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| | - Ignacio Fita
- Instituto
de Biologia Molecular de Barcelona (CSIC), Parc Científic de Barcelona, Baldiri i Reixac 10-12, 08028 Barcelona, Spain
| | - Ayush Kumar
- Department
of Microbiology, University of Manitoba, 45 Chancellor’s Circle, Winnipeg, Manitoba R3T 2N2, Canada
| |
Collapse
|
39
|
Karunaratne GHRE, Wijesundera SS, Vidanagama D, Adikaram CP, Perera J. Significance of Coexisting Mutations on Determination of the Degree of Isoniazid Resistance in Mycobacterium tuberculosis Strains. Microb Drug Resist 2018; 24:844-851. [PMID: 29683767 DOI: 10.1089/mdr.2017.0330] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The emergence and spread of drug-resistant tuberculosis (TB) pose a threat to TB control in Sri Lanka. Isoniazid (INH) is a key element of the first-line anti-TB treatment regimen. Resistance to INH is mainly associated with point mutations in katG, inhA, and ahpC genes. The objective of this study was to determine mutations of these three genes in INH-resistant Mycobacterium tuberculosis (MTb) strains in Sri Lanka. Complete nucleotide sequence of the three genes was amplified by polymerase chain reaction and subjected to DNA sequencing. Point mutations in the katG gene were identified in 93% isolates, of which the majority (78.6%) were at codon 315. Mutations at codons 212 and 293 of the katG gene have not been reported previously. Novel mutations were recognized in the promoter region of the inhA gene (C deletion at -34), fabG1 gene (codon 27), and ahpC gene (codon 39). Single S315T mutation in the katG gene led to a high level of resistance, while a low level of resistance with high frequency (41%) was observed when katG codon 315 coexisted with the mutation at codon 463. Since most of the observed mutations of all three genes coexisted with the katG315 mutation, screening of katG315 mutations will be a useful marker for molecular detection of INH resistance of MTb in Sri Lanka.
Collapse
Affiliation(s)
| | | | | | | | - Jennifer Perera
- 1 Department of Microbiology, Faculty of Medicine, University of Colombo , Colombo, Sri Lanka
| |
Collapse
|
40
|
Sharma K, Verma R, Advani J, Chatterjee O, Solanki HS, Sharma A, Varma S, Modi M, Ray P, Mukherjee KK, Sharma M, Dhillion MS, Suar M, Chatterjee A, Pandey A, Prasad TSK, Gowda H. Whole Genome Sequencing of Mycobacterium tuberculosis Isolates From Extrapulmonary Sites. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:413-425. [PMID: 28692415 DOI: 10.1089/omi.2017.0070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Tuberculosis (TB) remains one of the leading causes of morbidity and mortality worldwide. Extrapulmonary tuberculosis (EPTB) constitutes around 15-20% of TB cases in immunocompetent individuals. Extrapulmonary sites that are affected by TB include bones, lymph nodes, meningitis, pleura, and genitourinary tract. Whole genome sequencing has emerged as a powerful tool to map genetic diversity among Mycobacterium tuberculosis (MTB) isolates and identify the genomic signatures associated with drug resistance, pathogenesis, and disease transmission. Several pulmonary isolates of MTB have been sequenced over the years. However, availability of whole genome sequences of MTB isolates from extrapulmonary sites is limited. Some studies suggest that genetic variations in MTB might contribute to disease presentation in extrapulmonary sites. This can be addressed if whole genome sequence data from large number of extrapulmonary isolates becomes available. In this study, we have performed whole genome sequencing of five MTB clinical isolates derived from EPTB sites using next-generation sequencing platform. We identified 1434 nonsynonymous single nucleotide variations (SNVs), 143 insertions and 105 deletions. This includes 279 SNVs that were not reported before in publicly available datasets. We found several mutations that are known to confer resistance to drugs. All the five isolates belonged to East-African-Indian lineage (lineage 3). We identified 9 putative prophage DNA integrations and 14 predicted clustered regularly interspaced short palindromic repeats (CRISPR) in MTB genome. Our analysis indicates that more work is needed to map the genetic diversity of MTB. Whole genome sequencing in conjunction with comprehensive drug susceptibility testing can reveal clinically relevant mutations associated with drug resistance.
Collapse
Affiliation(s)
- Kusum Sharma
- 1 Department of Medical Microbiology, PGIMER , Chandigarh, India
| | - Renu Verma
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,3 School of Biotechnology, KIIT University , Bhubaneswar, India
| | - Jayshree Advani
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,4 Manipal University , Manipal, India
| | - Oishi Chatterjee
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,5 School of Biotechnology , Amrita Vishwa Vidyapeetham, Kollam, India
| | - Hitendra S Solanki
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,3 School of Biotechnology, KIIT University , Bhubaneswar, India
| | - Aman Sharma
- 6 Department of Internal Medicine, PGIMER, Chandigarh, India
| | - Subhash Varma
- 6 Department of Internal Medicine, PGIMER, Chandigarh, India
| | - Manish Modi
- 7 Department of Neurology, PGIMER, Chandigarh, India
| | - Pallab Ray
- 1 Department of Medical Microbiology, PGIMER , Chandigarh, India
| | | | - Megha Sharma
- 1 Department of Medical Microbiology, PGIMER , Chandigarh, India
| | | | - Mrutyunjay Suar
- 3 School of Biotechnology, KIIT University , Bhubaneswar, India
| | - Aditi Chatterjee
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,10 YU-IOB Center for Systems Biology and Molecular Medicine , Mangalore, India
| | - Akhilesh Pandey
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,11 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland.,12 Department of Biological Chemistry, Johns Hopkins University School of Medicine , Baltimore, Maryland.,13 Department of Pathology, Johns Hopkins University School of Medicine , Baltimore, Maryland.,14 Department of Oncology, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Thottethodi Subrahmanya Keshava Prasad
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,10 YU-IOB Center for Systems Biology and Molecular Medicine , Mangalore, India .,15 NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neurosciences , Bangalore, India
| | - Harsha Gowda
- 2 Institute of Bioinformatics , International Technology Park, Bangalore, India .,10 YU-IOB Center for Systems Biology and Molecular Medicine , Mangalore, India
| |
Collapse
|
41
|
Revisiting Activation of and Mechanism of Resistance to Compound IQG-607 in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2018; 62:AAC.02222-17. [PMID: 29158273 DOI: 10.1128/aac.02222-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/10/2017] [Indexed: 11/20/2022] Open
Abstract
IQG-607 is a metal complex previously reported as a promising anti-tuberculosis (TB) drug against isoniazid (INH)-resistant strains of Mycobacterium tuberculosis Unexpectedly, we found that INH-resistant clinical isolates were resistant to IQG-607. Spontaneous mutants resistant to IQG-607 were subjected to whole-genome sequencing, and all sequenced colonies carried alterations in the katG gene. The katG(S315T) mutation was sufficient to confer resistance to IQG-607 in both MIC assays and inside macrophages. Moreover, overexpression of the InhA(S94A) protein caused IQG-607's resistance.
Collapse
|
42
|
Vega-García V, Díaz-Vilchis A, Saucedo-Vázquez JP, Solano-Peralta A, Rudiño-Piñera E, Hansberg W. Structure, kinetics, molecular and redox properties of a cytosolic and developmentally regulated fungal catalase-peroxidase. Arch Biochem Biophys 2018; 640:17-26. [PMID: 29305053 DOI: 10.1016/j.abb.2017.12.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/24/2017] [Accepted: 12/29/2017] [Indexed: 11/18/2022]
Abstract
CAT-2, a cytosolic catalase-peroxidase (CP) from Neurospora crassa, which is induced during asexual spore formation, was heterologously expressed and characterized. CAT-2 had the Met-Tyr-Trp (M-Y-W) adduct required for catalase activity. Its KM for H2O2 was micromolar for peroxidase and millimolar for catalase activity. A Em = -158 mV reduction potential value was obtained and the Soret band shift suggested a mixture of low and high spin ferric iron. CAT-2 EPR spectrum at 10 K indicated an axial and a rhombic component. With peroxyacetic acid (PAA), formation of Compound I* was observed with EPR. CAT-2 homodimer crystallographic structure contained two K+ ions; Glu107 residues were displaced to bind them. CAT-2 showed the essential amino acid residues for activity in similar positions to other CPs. CAT-2 Arg426 is oriented towards the M-Y-W adduct, interacting with the deprotonated Tyr238 hydroxyl group. A perhydroxy modification of the indole nitrogen of Trp90 was oriented toward the catalytic His91. In contrast to cytochrome c peroxidase and ascorbate peroxidase, the catalase-peroxidase heme propionates are not exposed to the solvent. Together with other N. crassa enzymes that utilize H2O2 as a substrate, CAT-2 has many tryptophan and proline residues at its surface, probably related to H2O2 selection in water.
Collapse
Affiliation(s)
- Vanessa Vega-García
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Mexico
| | - Adelaida Díaz-Vilchis
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, Mexico
| | - Juan Pablo Saucedo-Vázquez
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Mexico
| | - Alejandro Solano-Peralta
- Unidad de Servicios de Apoyo a la Investigación y a la Industria, Facultad de Química, Universidad Nacional Autónoma de México, UNAM, Mexico
| | - Enrique Rudiño-Piñera
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, UNAM, Mexico
| | - Wilhelm Hansberg
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Mexico.
| |
Collapse
|
43
|
Purkan P, Ihsanawati I, Natalia D, Syah YM, Retnoningrum DS, Siswanto I. Molecular Analysis of katG Encoding Catalase-Peroxidase from Clinical Isolate of Isoniazid-Resistant Mycobacterium tuberculosis. J Med Life 2018; 11:160-167. [PMID: 30140323 PMCID: PMC6101688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Isoniazid (INH) is a drug for the treatment of tuberculosis in patients infected with Mycobacterium tuberculosis. The katG enzyme, or catalase-peroxidase, activates the pro-drug INH that is coded by the katG gene in M. tuberculosis. Mutations of the katG gene in M. tuberculosis are a major INH resistance mechanism. The M. tuberculosis clinical isolate R2 showed INH resistance at a high level of 10 µg/mL. However, the molecular basis for the resistance is unclear. The identification of a mutation in the katG gene of the clinical isolate R2 showed four mutations, i.e., C1061T, G1261 A, G1388T, G2161A, which correspond to the amino acid substitutions T354I, G421S, R463L, and V721M, respectively. The mutant katG gene, along with the wild-type were cloned, expressed and purified. The mutant enzyme showed 86.5% of catalase and 45% of peroxidase activities in comparison to the wild type. The substitutions of T354I and G421S in mutant katG R2 created significant instability in the adduct triad complex (Trp107-Tyr229-Met255), a part of the active site of the catalase-peroxidase enzyme in the model structure analysis. The events could be based on the high resistance of the clinical isolate R2 toward INH as the molecular basis.
Collapse
Affiliation(s)
- P Purkan
- Department of Chemistry, Faculty of Sciences and Technology, Airlangga University; Surabaya, Indonesia
| | - I Ihsanawati
- Biochemistry Research Division, Faculty of Mathematics and Natural Sciences, Bandung Institute of Technology, Bandung, Indonesia
| | - D Natalia
- Biochemistry Research Division, Faculty of Mathematics and Natural Sciences, Bandung Institute of Technology, Bandung, Indonesia
| | - YM Syah
- Biochemistry Research Division, Faculty of Mathematics and Natural Sciences, Bandung Institute of Technology, Bandung, Indonesia
| | - DS Retnoningrum
- School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - I Siswanto
- Department of Chemistry, Faculty of Sciences and Technology, Airlangga University; Surabaya, Indonesia
| |
Collapse
|
44
|
Ghazizadeh F, Ghaffari S, Mirshojaei SF, Mazidid M, Azarmi S. Biodistribution of Tc-99m Labeled Isoniazid Solid Lipid Nanoparticles in Wistar Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:1209-1216. [PMID: 30568681 PMCID: PMC6269554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
In this study Isoniazid (INH) as one of the first line drugs in treatment of Tuberculosis was investigated to be loaded in Solid Lipid Nanoparticles (SLNs) for reducing hepatotoxicity as well as prolonging drug release. High shear homogenization method was performed to prepare INH SLNs. To compare biodistribution of INH before and after loading in SLNs, INH was labeled by Technetium 99 (Tc99) after derivatization. The particle size of the prepared SLNs was 167 and 200 nm before and after lyophilization, respectively. Loading efficiency was calculated using the reverse method and release study was performed by using the dialysis sack method. Loading efficiency was 98%, and more than 85% of the loaded drug released in 3 h. Differential Scanning calorimeter (DSC) studies were performed for evaluating of the probability of happening hydrogen bonds or other chemical interactions between cholesterol as carrier and isoniazid as active pharmaceutical ingredient. The results could support the probability of hydrogen bond formation between cholesterol and INH. Gamma Scintigraphy studies showed that after administering INH SLNs, longer drug retention in the body was obtained compared to free INH. Quantitative gamma counting showed that the concentration of INH in the liver and intestines could be decreased by using nanotechnology.
Collapse
Affiliation(s)
- Fereshteh Ghazizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran.
| | - Solmaz Ghaffari
- Department of Medical Nanotechnology, Faculty of Advanced Technologies, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran. ,Young Researchers and Elite Club, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran. ,Pharmaceutical Sciences Research Center, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran. ,Corresponding author: E-mail:
| | - Seyedeh Fatemeh Mirshojaei
- Radiation Application Research School, Nuclear Science and Technology Research Institute, AEOI, Tehran, Iran.
| | - Mohammad Mazidid
- Radiation Application Research School, Nuclear Science and Technology Research Institute, AEOI, Tehran, Iran.
| | - Shirzad Azarmi
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alta, Canada.,Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
Laborde J, Deraeve C, Bernardes-Génisson V. Update of Antitubercular Prodrugs from a Molecular Perspective: Mechanisms of Action, Bioactivation Pathways, and Associated Resistance. ChemMedChem 2017; 12:1657-1676. [DOI: 10.1002/cmdc.201700424] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/12/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Julie Laborde
- CNRS; LCC (Laboratoire de Chimie de Coordination); 205, route de Narbonne, BP 44099 31077 Toulouse, Cedex 4 France
- Université de Toulouse; UPS, INPT; 31077 Toulouse, Cedex 4 France
| | - Céline Deraeve
- CNRS; LCC (Laboratoire de Chimie de Coordination); 205, route de Narbonne, BP 44099 31077 Toulouse, Cedex 4 France
- Université de Toulouse; UPS, INPT; 31077 Toulouse, Cedex 4 France
| | - Vania Bernardes-Génisson
- CNRS; LCC (Laboratoire de Chimie de Coordination); 205, route de Narbonne, BP 44099 31077 Toulouse, Cedex 4 France
- Université de Toulouse; UPS, INPT; 31077 Toulouse, Cedex 4 France
| |
Collapse
|
46
|
Njuma OJ, Davis I, Ndontsa EN, Krewall JR, Liu A, Goodwin DC. Mutual synergy between catalase and peroxidase activities of the bifunctional enzyme KatG is facilitated by electron hole-hopping within the enzyme. J Biol Chem 2017; 292:18408-18421. [PMID: 28972181 DOI: 10.1074/jbc.m117.791202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/22/2017] [Indexed: 11/06/2022] Open
Abstract
KatG is a bifunctional, heme-dependent enzyme in the front-line defense of numerous bacterial and fungal pathogens against H2O2-induced oxidative damage from host immune responses. Contrary to the expectation that catalase and peroxidase activities should be mutually antagonistic, peroxidatic electron donors (PxEDs) enhance KatG catalase activity. Here, we establish the mechanism of synergistic cooperation between these activities. We show that at low pH values KatG can fully convert H2O2 to O2 and H2O only if a PxED is present in the reaction mixture. Stopped-flow spectroscopy results indicated rapid initial rates of H2O2 disproportionation slowing concomitantly with the accumulation of ferryl-like heme states. These states very slowly returned to resting (i.e. ferric) enzyme, indicating that they represented catalase-inactive intermediates. We also show that an active-site tryptophan, Trp-321, participates in off-pathway electron transfer. A W321F variant in which the proximal tryptophan was replaced with a non-oxidizable phenylalanine exhibited higher catalase activity and less accumulation of off-pathway heme intermediates. Finally, rapid freeze-quench EPR experiments indicated that both WT and W321F KatG produce the same methionine-tyrosine-tryptophan (MYW) cofactor radical intermediate at the earliest reaction time points and that Trp-321 is the preferred site of off-catalase protein oxidation in the native enzyme. Of note, PxEDs did not affect the formation of the MYW cofactor radical but could reduce non-productive protein-based radical species that accumulate during reaction with H2O2 Our results suggest that catalase-inactive intermediates accumulate because of off-mechanism oxidation, primarily of Trp-321, and PxEDs stimulate KatG catalase activity by preventing the accumulation of inactive intermediates.
Collapse
Affiliation(s)
- Olive J Njuma
- From the Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849-5312
| | - Ian Davis
- the Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249-0698, and.,the Department of Chemistry, Georgia State University, Atlanta, Georgia 30303
| | - Elizabeth N Ndontsa
- From the Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849-5312
| | - Jessica R Krewall
- From the Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849-5312
| | - Aimin Liu
- the Department of Chemistry, University of Texas at San Antonio, San Antonio, Texas 78249-0698, and
| | - Douglas C Goodwin
- From the Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849-5312,
| |
Collapse
|
47
|
Gönlügür T, Gönlügür U. INH Direnç Mekanizmaları. DICLE MEDICAL JOURNAL 2017. [DOI: 10.5798/dicletip.339020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
48
|
Lone MY, Athar M, Gupta VK, Jha PC. Identification of Mycobacterium tuberculosis enoyl-acyl carrier protein reductase inhibitors: A combined in-silico and in-vitro analysis. J Mol Graph Model 2017; 76:172-180. [DOI: 10.1016/j.jmgm.2017.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/20/2023]
|
49
|
Abstract
The tuberculosis agent Mycobacterium tuberculosis has undergone a long and selective evolution toward human infection and represents one of the most widely spread pathogens due to its efficient aerosol-mediated human-to-human transmission. With the availability of more and more genome sequences, the evolutionary trajectory of this obligate pathogen becomes visible, which provides us with new insights into the molecular events governing evolution of the bacterium and its ability to accumulate drug-resistance mutations. In this review, we summarize recent developments in mycobacterial research related to this matter that are important for a better understanding of the current situation and future trends and developments in the global epidemiology of tuberculosis, as well as for possible public health intervention possibilities.
Collapse
|
50
|
Manson AL, Abeel T, Galagan JE, Sundaramurthi JC, Salazar A, Gehrmann T, Shanmugam SK, Palaniyandi K, Narayanan S, Swaminathan S, Earl AM. Mycobacterium tuberculosis Whole Genome Sequences From Southern India Suggest Novel Resistance Mechanisms and the Need for Region-Specific Diagnostics. Clin Infect Dis 2017; 64:1494-1501. [PMID: 28498943 PMCID: PMC5434337 DOI: 10.1093/cid/cix169] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/30/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND. India is home to 25% of all tuberculosis cases and the second highest number of multidrug resistant cases worldwide. However, little is known about the genetic diversity and resistance determinants of Indian Mycobacterium tuberculosis, particularly for the primary lineages found in India, lineages 1 and 3. METHODS. We whole genome sequenced 223 randomly selected M. tuberculosis strains from 196 patients within the Tiruvallur and Madurai districts of Tamil Nadu in Southern India. Using comparative genomics, we examined genetic diversity, transmission patterns, and evolution of resistance. RESULTS. Genomic analyses revealed (11) prevalence of strains from lineages 1 and 3, (11) recent transmission of strains among patients from the same treatment centers, (11) emergence of drug resistance within patients over time, (11) resistance gained in an order typical of strains from different lineages and geographies, (11) underperformance of known resistance-conferring mutations to explain phenotypic resistance in Indian strains relative to studies focused on other geographies, and (11) the possibility that resistance arose through mutations not previously implicated in resistance, or through infections with multiple strains that confound genotype-based prediction of resistance. CONCLUSIONS. In addition to substantially expanding the genomic perspectives of lineages 1 and 3, sequencing and analysis of M. tuberculosis whole genomes from Southern India highlight challenges of infection control and rapid diagnosis of resistant tuberculosis using current technologies. Further studies are needed to fully explore the complement of diversity and resistance determinants within endemic M. tuberculosis populations.
Collapse
Affiliation(s)
| | - Thomas Abeel
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Delft Bioinformatics Lab, Delft University of Technology, The Netherlands
| | - James E Galagan
- Department of Biomedical Engineering, and
- National Emerging Infectious Diseases Laboratory, Boston University, Massachusetts
| | | | - Alex Salazar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Delft Bioinformatics Lab, Delft University of Technology, The Netherlands
| | - Thies Gehrmann
- Delft Bioinformatics Lab, Delft University of Technology, The Netherlands
| | | | | | | | | | - Ashlee M Earl
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|