151
|
Mucaji P, Atanasov AG, Bak A, Kozik V, Sieron K, Olsen M, Pan W, Liu Y, Hu S, Lan J, Haider N, Musiol R, Vanco J, Diederich M, Ji S, Zitko J, Wang D, Agbaba D, Nikolic K, Oljacic S, Vucicevic J, Jezova D, Tsantili-Kakoulidou A, Tsopelas F, Giaginis C, Kowalska T, Sajewicz M, Silberring J, Mielczarek P, Smoluch M, Jendrzejewska I, Polanski J, Jampilek J. The Forty-Sixth Euro Congress on Drug Synthesis and Analysis: Snapshot †. Molecules 2017; 22:molecules22111848. [PMID: 29143778 PMCID: PMC6150335 DOI: 10.3390/molecules22111848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
The 46th EuroCongress on Drug Synthesis and Analysis (ECDSA-2017) was arranged within the celebration of the 65th Anniversary of the Faculty of Pharmacy at Comenius University in Bratislava, Slovakia from 5-8 September 2017 to get together specialists in medicinal chemistry, organic synthesis, pharmaceutical analysis, screening of bioactive compounds, pharmacology and drug formulations; promote the exchange of scientific results, methods and ideas; and encourage cooperation between researchers from all over the world. The topic of the conference, "Drug Synthesis and Analysis," meant that the symposium welcomed all pharmacists and/or researchers (chemists, analysts, biologists) and students interested in scientific work dealing with investigations of biologically active compounds as potential drugs. The authors of this manuscript were plenary speakers and other participants of the symposium and members of their research teams. The following summary highlights the major points/topics of the meeting.
Collapse
Affiliation(s)
- Pavel Mucaji
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia.
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Andrzej Bak
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Violetta Kozik
- Department of Synthesis Chemistry, Faculty of Mathematics, Physics and Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Karolina Sieron
- Department of Physical Medicine, Medical University of Silesia, Medykow 18, 40752 Katowice, Poland.
| | - Mark Olsen
- Department of Pharmaceutical Sciences, College of Pharmacy Glendale, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA.
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Yazhou Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Shengchao Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Junjie Lan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, 3491 Baijin Road, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, 3491 Baijin Road, Guiyang, 550014, China.
| | - Norbert Haider
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstraße 14, A-1090 Vienna, Austria.
| | - Robert Musiol
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Jan Vanco
- Department of Inorganic Chemistry & Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University, 17. listopadu 12, 77146 Olomouc, Czech Republic.
| | - Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Seoul 08826, Korea.
| | - Seungwon Ji
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Seoul 08826, Korea.
| | - Jan Zitko
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic.
| | - Dongdong Wang
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Postepu 36A, 05-552 Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Danica Agbaba
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Jelica Vucicevic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Daniela Jezova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia.
| | - Anna Tsantili-Kakoulidou
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece.
| | - Fotios Tsopelas
- Laboratory of Inorganic and Analytical Chemistry, School of Chemical Engineering, National Technical University of Athens, Iroon Polytechniou 9, 15780 Athens, Greece.
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece.
| | - Teresa Kowalska
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Mieczyslaw Sajewicz
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Jerzy Silberring
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Przemyslaw Mielczarek
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Marek Smoluch
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University of Science and Technology, Mickiewicza 30, 30059 Krakow, Poland.
| | - Izabela Jendrzejewska
- Department of Crystallography, Faculty of Mathematics, Physics and Chemistry, University of Silesia, Bankowa 12, 40006 Katowice, Poland.
| | - Jaroslaw Polanski
- Institute of Chemistry, University of Silesia, Szkolna 9, 40007 Katowice, Poland.
| | - Josef Jampilek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Comenius University, Odbojarov 10, 83232 Bratislava, Slovakia.
| |
Collapse
|
152
|
Ureidopyrazine Derivatives: Synthesis and Biological Evaluation as Anti-Infectives and Abiotic Elicitors. Molecules 2017; 22:molecules22101797. [PMID: 29065539 PMCID: PMC6151446 DOI: 10.3390/molecules22101797] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 11/17/2022] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) has become a frequently deadly infection due to increasing antimicrobial resistance. This serious issue has driven efforts worldwide to discover new drugs effective against Mtb. One research area is the synthesis and evaluation of pyrazinamide derivatives as potential anti-TB drugs. In this paper we report the synthesis and biological evaluations of a series of ureidopyrazines. Compounds were synthesized by reacting alkyl/aryl isocyanates with aminopyrazine or with propyl 5-aminopyrazine-2-carboxylate. Reactions were performed in pressurized vials using a CEM Discover microwave reactor with a focused field. Purity and chemical structures of products were assessed, and the final compounds were tested in vitro for their antimycobacterial, antibacterial, and antifungal activities. Propyl 5-(3-phenylureido)pyrazine-2-carboxylate (compound 4, MICMtb = 1.56 μg/mL, 5.19 μM) and propyl 5-(3-(4-methoxyphenyl)ureido)pyrazine-2-carboxylate (compound 6, MICMtb = 6.25 μg/mL, 18.91 μM) had high antimycobacterial activity against Mtb H37Rv with no in vitro cytotoxicity on HepG2 cell line. Therefore 4 and 6 are suitable for further structural modifications that might improve their biological activity and physicochemical properties. Based on the structural similarity to 1-(2-chloropyridin-4-yl)-3-phenylurea, a known plant growth regulator, two selected compounds were evaluated for similar activity as abiotic elicitors.
Collapse
|
153
|
Molecular analysis of pyrazinamide resistance in Mycobacterium tuberculosis in Vietnam highlights the high rate of pyrazinamide resistance-associated mutations in clinical isolates. Emerg Microbes Infect 2017; 6:e86. [PMID: 29018250 PMCID: PMC5658769 DOI: 10.1038/emi.2017.73] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/16/2017] [Accepted: 08/06/2017] [Indexed: 11/25/2022]
Abstract
Pyrazinamide (PZA) is a key antibiotic in current anti-tuberculosis regimens. Although the WHO has stressed the urgent need to obtain data on PZA resistance, in high tuberculosis burden countries, little is known about the level of PZA resistance, the genetic basis of such resistance or its link with Mycobacterium tuberculosis families. In this context, this study assessed PZA resistance through the molecular analysis of 260 Vietnamese M. tuberculosis isolates. First-line drug susceptibility testing, pncA gene sequencing, spoligotyping and mycobacterial interspersed repetitive units-variable number of tandem repeats (MIRU-VNTR) typing were performed. Overall, the pncA mutation frequency was 38.1% (99 out of 260 isolates) but was higher than 72% (89 out of 123 isolates) in multidrug and quadruple-drug resistant isolates. Many different pncA mutations (71 types) were detected, of which 55 have been previously described and 50 were linked to PZA resistance. Among the 16 novel mutations, 14 are likely to be linked to PZA resistance because of their mutation types or codon positions. Genotype analysis revealed that PZA resistance can emerge in any M. tuberculosis cluster or family, although the mutation frequency was the highest in Beijing family isolates (47.7%, 62 out of 130 isolates). These data highlight the high rate of PZA resistance-associated mutations in M. tuberculosis clinical isolates in Vietnam and bring into question the use of PZA for current and future treatment regimens of multidrug-resistant tuberculosis without PZA resistance testing.
Collapse
|
154
|
Sheen P, Requena D, Gushiken E, Gilman RH, Antiparra R, Lucero B, Lizárraga P, Cieza B, Roncal E, Grandjean L, Pain A, McNerney R, Clark TG, Moore D, Zimic M. A multiple genome analysis of Mycobacterium tuberculosis reveals specific novel genes and mutations associated with pyrazinamide resistance. BMC Genomics 2017; 18:769. [PMID: 29020922 PMCID: PMC5637355 DOI: 10.1186/s12864-017-4146-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 10/02/2017] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Tuberculosis (TB) is a major global health problem and drug resistance compromises the efforts to control this disease. Pyrazinamide (PZA) is an important drug used in both first and second line treatment regimes. However, its complete mechanism of action and resistance remains unclear. RESULTS We genotyped and sequenced the complete genomes of 68 M. tuberculosis strains isolated from unrelated TB patients in Peru. No clustering pattern of the strains was verified based on spoligotyping. We analyzed the association between PZA resistance with non-synonymous mutations and specific genes. We found mutations in pncA and novel genes significantly associated with PZA resistance in strains without pncA mutations. These included genes related to transportation of metal ions, pH regulation and immune system evasion. CONCLUSIONS These results suggest potential alternate mechanisms of PZA resistance that have not been found in other populations, supporting that the antibacterial activity of PZA may hit multiple targets.
Collapse
Affiliation(s)
- Patricia Sheen
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - David Requena
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Eduardo Gushiken
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Robert H. Gilman
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe St., Room 5515, Baltimore, MD 21205 USA
| | - Ricardo Antiparra
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Bryan Lucero
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Pilar Lizárraga
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Basilio Cieza
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Elisa Roncal
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| | - Louis Grandjean
- Department of Infection, Immunology and Rheumatology, Institute of Child Health, University College London, 30 Guilford St, London, WC1N 1EH UK
| | - Arnab Pain
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | - Ruth McNerney
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT UK
| | - Taane G. Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT UK
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, WC1E 7HT UK
| | - David Moore
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT UK
| | - Mirko Zimic
- Laboratorio de Bioinformática y Biología Molecular. Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porras, 31 Lima, Peru
| |
Collapse
|
155
|
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: 3.0] [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
|
156
|
Yang J, Liu Y, Liu Z, Meng C, Lin D. Backbone and side-chain resonance assignments for the tmRNA-binding protein, SmpB, from Mycobacterium tuberculosis. BIOMOLECULAR NMR ASSIGNMENTS 2017; 11:175-179. [PMID: 28258549 DOI: 10.1007/s12104-017-9742-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 02/24/2017] [Indexed: 06/06/2023]
Abstract
Small protein B (SmpB) is an essential molecule in trans-translation which is a universal biological pathway for protein synthesis in bacteria. Trans-translation can release stalled ribosomes from defective mRNAs and target tag-protein fragments for degradation, and then restart protein synthesis. The SmpB-tmRNA complex coordinating with other components of the trans-translation system, plays vital roles in Mycobacterium tuberculosis under both stress conditions and non-replicating conditions. Thus, elucidation of molecular details and dynamic properties of the SmpB-tmRNA interaction is a crucial step towards effectively blocking trans-translation process to shorten the duration of tuberculosis treatment. Here, we report resonance assignments for 1H, 13C and 15N of M. tuberculosis SmpB (MtSmpB, spanning residues 4-133) protein determined by a suite of 2D/3D heteronuclear NMR experiments along with predicted the secondary structure.
Collapse
Affiliation(s)
- Juanjuan Yang
- Institute of Pharmaceutical Biotechnology and Engineering, College of Biological Science and Biotechnology, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Yindi Liu
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Zhao Liu
- Institute of Pharmaceutical Biotechnology and Engineering, College of Biological Science and Biotechnology, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Chun Meng
- Institute of Pharmaceutical Biotechnology and Engineering, College of Biological Science and Biotechnology, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Donghai Lin
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
157
|
Drug development against tuberculosis: Past, present and future. ACTA ACUST UNITED AC 2017; 64:252-275. [DOI: 10.1016/j.ijtb.2017.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/15/2017] [Indexed: 12/29/2022]
|
158
|
Fu J, Huang B, Lin D, Liao X. Chemical shift assignments of Ribosomal protein S1 from Mycobacterium tuberculosis. BIOMOLECULAR NMR ASSIGNMENTS 2017; 11:133-136. [PMID: 28258550 DOI: 10.1007/s12104-017-9734-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/20/2017] [Indexed: 06/06/2023]
Abstract
RpsA, also known as ribosomal protein S1, is an essential protein required for translation initiation of mRNAs when their Shine-Dalgarno sequence is degenerated (Sorensen et al. 1998). In addition, RpsA of Mycobacterium tuberculosis (M. tb) is involved in trans-translation, which is an effective system mediated by tmRNA-SmpB to release stalled ribosomes from mRNA in the presence of rare codons (Keiler 2008). Shi et al. found that POA binds to RpsA of Mtb and disrupts the formation of RpsA-tmRNA complex (Shi et al. 2011) and mutations at the C-terminus of RpsA confer PZA resistance. The previous work reported the pyrazinoic acid-binding domain of RpsA (Yang et al. Mol Microbiol 95:791-803, 2015). However, the HSQC spectra of the isolated S1 domain does not overlap with that of MtRpsA280-438, suggesting that substantial interactions occur between the flexible C-terminus and the S1 domain in MtRpsA .To further study the PZA resistance and how substantial interactions influence/affect protein structure, using heteronuclear NMR spectroscopy, we have completed backbone and side-chain 1H, 15N, 13C chemical shift assignments of MtRpsA280-438 which contains S1 domain and the flexible C-terminus. These NMR resonance assignments provide the framework for detailed characterization of the solution-state protein structure determination, dynamic studies of this domain, as well as NMR-based drug discovery efforts.
Collapse
Affiliation(s)
- Jinglin Fu
- High-field NMR Research Center, Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Biling Huang
- High-field NMR Research Center, Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Donghai Lin
- High-field NMR Research Center, Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Xinli Liao
- High-field NMR Research Center, Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
159
|
Al-Humadi HW, Al-Saigh RJ, Al-Humadi AW. Addressing the Challenges of Tuberculosis: A Brief Historical Account. Front Pharmacol 2017; 8:689. [PMID: 29033842 PMCID: PMC5626940 DOI: 10.3389/fphar.2017.00689] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/14/2017] [Indexed: 02/04/2023] Open
Abstract
Tuberculosis (TB) is a highly contagious disease that still poses a threat to human health. Mycobacterium tuberculosis (MTB), the pathogen responsible for TB, uses diverse ways in order to survive in a variety of host lesions and to subsequently evade immune surveillance; as a result, fighting TB and its associated multidrug resistance has been an ongoing challenge. The aim of this review article is to summarize the historical sequence of drug development and use in the fight against TB, with a particular emphasis on the decades between World War II and the dawn of the twenty first century (2000).
Collapse
Affiliation(s)
- Hussam W. Al-Humadi
- Department of Pharmacology and Toxicology, Pharmacy College, University of Babylon, Babylon, Iraq
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Rafal J. Al-Saigh
- Department of Pharmacology and Toxicology, Pharmacy College, University of Babylon, Babylon, Iraq
| | - Ahmed W. Al-Humadi
- Laboratory of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
160
|
Biofilms: Survival and defense strategy for pathogens. Int J Med Microbiol 2017; 307:481-489. [PMID: 28950999 DOI: 10.1016/j.ijmm.2017.09.016] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 01/20/2023] Open
Abstract
Studies on biofilm related infections are gaining prominence owing to their involvement in majority of clinical infections. Biofilm, considered as a generic mechanism for survival used by pathogenic as well as non-pathogenic microorganisms, involves surface attachment and growth of heterogeneous cells encapsulated within a matrix. The matrix provides ecological niche where partnership of cells endows a community like behaviour that not only enables the cohort to survive local microenvironment stress but also channelizes them to evolve, disseminate and cause resurgence of infections. In this mini-review we highlight the mechanisms used by microbes to develop and sustain biofilms, including the influence of the microbiota. Several strategies to target biofilms have been validated on certain groups of microorganisms and these basically target different stages in the life cycle of biofilm, however comprehensive methods to target microbial biofilms are relatively unknown. In the backdrop of recent reports suggesting that biofilms can harbour multiple species of organisms, we need to relook and devise newer strategies against biofilms. Effective anti-biofilm strategies cannot be confined to a single methodology that can disrupt one pathway but should simultaneously target the various routes adopted by the microorganisms for survival within their ecosystem. An overview of the currently available drugs, their mode of action, genomic targets and translational therapies against biofilm related infection are discussed.
Collapse
|
161
|
Yadon AN, Maharaj K, Adamson JH, Lai YP, Sacchettini JC, Ioerger TR, Rubin EJ, Pym AS. A comprehensive characterization of PncA polymorphisms that confer resistance to pyrazinamide. Nat Commun 2017; 8:588. [PMID: 28928454 PMCID: PMC5605632 DOI: 10.1038/s41467-017-00721-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/24/2017] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis chemotherapy is dependent on the use of the antibiotic pyrazinamide, which is being threatened by emerging drug resistance. Resistance is mediated through mutations in the bacterial gene pncA. Methods for testing pyrazinamide susceptibility are difficult and rarely performed, and this means that the full spectrum of pncA alleles that confer clinical resistance to pyrazinamide is unknown. Here, we performed in vitro saturating mutagenesis of pncA to generate a comprehensive library of PncA polymorphisms resultant from a single-nucleotide polymorphism. We then screened it for pyrazinamide resistance both in vitro and in an infected animal model. We identify over 300 resistance-conferring substitutions. Strikingly, these mutations map throughout the PncA structure and result in either loss of enzymatic activity and/or decrease in protein abundance. Our comprehensive mutational and screening approach should stand as a paradigm for determining resistance mutations and their mechanisms of action.The antibiotic pyrazinamide is central to tuberculosis treatment regimens, globally. Despite its efficacy, resistance to the drug is increasing. Here, Eric Rubin and colleagues characterise the genetic basis of pyrazinamide resistance.
Collapse
Affiliation(s)
- Adam N Yadon
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, 665 Huntington Ave., Bldg 1, Rm 810, Boston, MA, 02115, USA
- African Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, K-RITH Tower Building, Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Kashmeel Maharaj
- African Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, K-RITH Tower Building, Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - John H Adamson
- African Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, K-RITH Tower Building, Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Yi-Pin Lai
- Department of Computer Science and Engineering, 3112 Texas A&M University, 301 H.R. Bright Building, College Station, TX, 77843, USA
| | - James C Sacchettini
- Department of Biochemistry and Biophysics, Texas A&M University, Interdisciplinary Life Sciences Building, 301 Old Main Dr., College Station, TX, 77843, USA
| | - Thomas R Ioerger
- Department of Computer Science and Engineering, 3112 Texas A&M University, 301 H.R. Bright Building, College Station, TX, 77843, USA
| | - Eric J Rubin
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, 665 Huntington Ave., Bldg 1, Rm 810, Boston, MA, 02115, USA.
| | - Alexander S Pym
- African Health Research Institute (AHRI), Nelson R. Mandela School of Medicine, K-RITH Tower Building, Level 3, 719 Umbilo Road, Durban, 4001, South Africa.
| |
Collapse
|
162
|
Alumasa JN, Manzanillo PS, Peterson ND, Lundrigan T, Baughn AD, Cox JS, Keiler KC. Ribosome Rescue Inhibitors Kill Actively Growing and Nonreplicating Persister Mycobacterium tuberculosis Cells. ACS Infect Dis 2017; 3:634-644. [PMID: 28762275 PMCID: PMC5594445 DOI: 10.1021/acsinfecdis.7b00028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
The
emergence of Mycobacterium tuberculosis (MTB) strains
that are resistant to most or all available antibiotics has created
a severe problem for treating tuberculosis and has spurred a quest
for new antibiotic targets. Here, we demonstrate that trans-translation is essential for growth of MTB and is a viable target
for development of antituberculosis drugs. We also show that an inhibitor
of trans-translation, KKL-35, is bactericidal against
MTB under both aerobic and anoxic conditions. Biochemical experiments
show that this compound targets helix 89 of the 23S rRNA. In silico molecular docking predicts a binding pocket for
KKL-35 adjacent to the peptidyl-transfer center in a region not targeted
by conventional antibiotics. Computational solvent mapping suggests
that this pocket is a druggable hot spot for small molecule binding.
Collectively, our findings reveal a new target for antituberculosis
drug development and provide critical insight on the mechanism of
antibacterial action for KKL-35 and related 1,3,4-oxadiazole benzamides.
Collapse
Affiliation(s)
- John N. Alumasa
- Department of Biochemistry
and Molecular Biology, The Pennsylvania State University, 401 Althouse Laboratory, University Park, Pennsylvania 16802, United States
| | - Paolo S. Manzanillo
- Department
of Molecular and Cell Biology, University of California, Berkeley, #3370, 375E Li Ka Shing Center, Berkeley, California 94720, United States
| | - Nicholas D. Peterson
- Department of Microbiology and Immunology,
Microbiology Research Facility, University of Minnesota, Rm4-115, 689 23rd Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Tricia Lundrigan
- Department
of Molecular and Cell Biology, University of California, Berkeley, #3370, 375E Li Ka Shing Center, Berkeley, California 94720, United States
| | - Anthony D. Baughn
- Department of Microbiology and Immunology,
Microbiology Research Facility, University of Minnesota, Rm4-115, 689 23rd Ave. SE, Minneapolis, Minnesota 55455, United States
| | - Jeffery S. Cox
- Department
of Molecular and Cell Biology, University of California, Berkeley, #3370, 375E Li Ka Shing Center, Berkeley, California 94720, United States
| | - Kenneth C. Keiler
- Department of Biochemistry
and Molecular Biology, The Pennsylvania State University, 401 Althouse Laboratory, University Park, Pennsylvania 16802, United States
| |
Collapse
|
163
|
Design, Synthesis, Antimycobacterial Evaluation, and In Silico Studies of 3-(Phenylcarbamoyl)-pyrazine-2-carboxylic Acids. Molecules 2017; 22:molecules22091491. [PMID: 28880230 PMCID: PMC6151461 DOI: 10.3390/molecules22091491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 09/05/2017] [Indexed: 11/17/2022] Open
Abstract
Pyrazinamide, the first-line antitubercular drug, has been regarded the basic component of tuberculosis treatment for over sixty years. Researchers have investigated its effect on Mycobacterium tuberculosis for this long time, and as a result, new potential targets of pyrazinamide or its active form, pyrazinoic acid, have been found. We have designed and prepared 3-(phenyl-carbamoyl)pyrazine-2-carboxylic acids as more lipophilic derivatives of pyrazinoic acid. We also prepared methyl and propyl derivatives as prodrugs with further increased lipophilicity. Antimycobacterial, antibacterial and antifungal growth inhibiting activity was investigated in all prepared compounds. 3-[(4-Nitrophenyl)carbamoyl]pyrazine-2-carboxylic acid (16) exerted high antimycobacterial activity against Mycobacterium tuberculosis H37Rv with MIC = 1.56 μg·mL−1 (5 μM). Propyl 3-{[4-(trifluoromethyl)phenyl]carbamoyl}pyrazine-2-carboxylate (18a) showed also high antimycobacterial activity against Mycobacterium tuberculosis H37Rv with MIC = 3.13 μg·mL−1. In vitro cytotoxicity of the active compounds was investigated and no significant cytotoxic effect was observed. Based to structural similarity to known inhibitors of decaprenylphosphoryl-β-d-ribose oxidase, DprE1, we performed molecular docking of the prepared acids to DprE1. These in silico experiments indicate that modification of the linker connecting aromatic parts of molecule does not have any negative influence on the binding.
Collapse
|
164
|
Mishra SK, Tripathi G, Kishore N, Singh RK, Singh A, Tiwari VK. Drug development against tuberculosis: Impact of alkaloids. Eur J Med Chem 2017. [DOI: 10.1016/j.ejmech.2017.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
165
|
Larsen EM, Stephens DC, Clarke NH, Johnson RJ. Ester-prodrugs of ethambutol control its antibacterial activity and provide rapid screening for mycobacterial hydrolase activity. Bioorg Med Chem Lett 2017; 27:4544-4547. [PMID: 28882482 DOI: 10.1016/j.bmcl.2017.08.057] [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: 05/30/2017] [Revised: 08/24/2017] [Accepted: 08/27/2017] [Indexed: 10/19/2022]
Abstract
M. tuberculosis contains an unusually high number of serine hydrolases by proteome percentage compared to other common bacteria or humans. This letter describes a method to probe the global substrate specificity of mycobacterial serine hydrolases with ester-protected prodrugs of ethambutol, a first-line antibiotic treatment for TB. These compounds were synthesized directly from ethambutol using a selective o-acylation to yield products in high yield and purity with minimal workup. A library of derivatives was screened against M. smegmatis, a non-infectious model for M. tuberculosis, which displayed significantly lowered biological activity compared to ethambutol. Incubation with a general serine hydrolase reactivated each derivative to near-ethambutol levels, demonstrating that esterification of ethambutol should provide a simple screen for mycobacterial hydrolase activity.
Collapse
Affiliation(s)
- Erik M Larsen
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave., Indianapolis, IN 46208, USA
| | - Dominique C Stephens
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave., Indianapolis, IN 46208, USA
| | - Nathan H Clarke
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave., Indianapolis, IN 46208, USA
| | - R Jeremy Johnson
- Department of Chemistry and Biochemistry, Butler University, 4600 Sunset Ave., Indianapolis, IN 46208, USA.
| |
Collapse
|
166
|
Pyrazinamide Susceptibility and pncA Mutation Profiles of Mycobacterium tuberculosis among Multidrug-Resistant Tuberculosis Patients in Bangladesh. Antimicrob Agents Chemother 2017. [PMID: 28630193 PMCID: PMC5571327 DOI: 10.1128/aac.00511-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pyrazinamide (PZA) is a frontline antituberculosis (anti-TB) drug used in both first- and second-line treatment regimens. However, due to complex laboratory requirements, the PZA susceptibility test is rarely performed, leading to a scarcity of data on susceptibility to PZA. Bangladesh is a country with a burden of high rates of both TB and multidrug-resistant TB (MDR-TB), but to our knowledge, published data on rates of PZA susceptibility (PZAs), especially among MDR-TB patients, are limited. We aimed to analyze the PZA susceptibility patterns of Mycobacterium tuberculosis isolates from MDR-TB patients and to correlate the pncA mutation with PZA resistance in Bangladesh. A total of 169 confirmed MDR M. tuberculosis isolates from a pool of specimens collected in a nationwide surveillance study were included in this analysis. All the isolates were tested for phenotypic PZA susceptibility in Bactec mycobacterial growth indicator tube (MGIT) culture medium, and the pncA gene was sequenced. We also correlated different types of clinical information and treatment outcomes with PZA susceptibility. We found that 45% of isolates were phenotypically PZA resistant. Sequencing of the pncA gene revealed a high concordance (82.2%) between the pncA gene sequence and the phenotypic assay results. A total of 64 different mutations were found, and 9 isolates harbored multiple mutations. We detected 27 new pncA mutations. We did not find any significant correlation between the different clinical categories, the genetic lineage, or treatment outcome group and PZA susceptibility. Considering the turnaround time, sequencing would be the more feasible option to determine PZA susceptibility, and further studies to investigate the MIC of PZA should be conducted to determine an effective dose of the drug.
Collapse
|
167
|
Identification of Novel Efflux Proteins Rv0191, Rv3756c, Rv3008, and Rv1667c Involved in Pyrazinamide Resistance in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2017; 61:AAC.00940-17. [PMID: 28584158 DOI: 10.1128/aac.00940-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 05/28/2017] [Indexed: 11/20/2022] Open
Abstract
Pyrazinamide (PZA) is a critical drug used for the treatment of tuberculosis (TB). PZA is a prodrug that requires conversion to the active component pyrazinoic acid (POA) by pyrazinamidase (PZase) encoded by the pncA gene. Although resistance to PZA is mostly caused by pncA mutations and less commonly by rpsA, panD, and clpC1 mutations, clinical strains without these mutations are known to exist. While efflux of POA was demonstrated in Mycobacterium tuberculosis previously, the efflux proteins involved have not been identified. Here we performed POA binding studies with an M. tuberculosis proteome microarray and identified four efflux proteins (Rv0191, Rv3756c, Rv3008, and Rv1667c) that bind POA. Overexpression of the four efflux pump genes in M. tuberculosis caused low-level resistance to PZA and POA but not to other drugs. Furthermore, addition of efflux pump inhibitors such as reserpine, piperine, and verapamil caused increased susceptibility to PZA in M. tuberculosis strains overexpressing the efflux proteins Rv0191, Rv3756c, Rv3008, and Rv1667c. Our studies indicate that these four efflux proteins may be responsible for PZA/POA efflux and cause PZA resistance in M. tuberculosis Future studies are needed to assess their roles in PZA resistance in clinical strains.
Collapse
|
168
|
Dillon NA, Peterson ND, Feaga HA, Keiler KC, Baughn AD. Anti-tubercular Activity of Pyrazinamide is Independent of trans-Translation and RpsA. Sci Rep 2017; 7:6135. [PMID: 28733601 PMCID: PMC5522395 DOI: 10.1038/s41598-017-06415-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022] Open
Abstract
Pyrazinamide (PZA) is a first line anti-tubercular drug for which the mechanism of action remains unresolved. Recently, it was proposed that the active form of PZA, pyrazinoic acid (POA), disrupts the ribosome rescue process of trans-translation in Mycobacterium tuberculosis. This model suggested that POA binds within the carboxy-terminal domain of ribosomal protein S1 (RpsA) and inhibits trans-translation leading to accumulation of stalled ribosomes. Here, we demonstrate that M. tuberculosis RpsA interacts with single stranded RNA, but not with POA. Further, we show that an rpsA polymorphism previously identified in a PZA resistant strain does not confer PZA resistance when reconstructed in a laboratory strain. Finally, by utilizing an in vitro trans-translation assay with purified M. tuberculosis ribosomes we find that an interfering oligonucleotide can inhibit trans-translation, yet POA does not inhibit trans-translation. Based on these findings, we conclude that the action of PZA is entirely independent of RpsA and trans-translation in M. tuberculosis.
Collapse
Affiliation(s)
- Nicholas A Dillon
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Nicholas D Peterson
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Heather A Feaga
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kenneth C Keiler
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Anthony D Baughn
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
169
|
Gopal P, Tasneen R, Yee M, Lanoix JP, Sarathy J, Rasic G, Li L, Dartois V, Nuermberger E, Dick T. In Vivo-Selected Pyrazinoic Acid-Resistant Mycobacterium tuberculosis Strains Harbor Missense Mutations in the Aspartate Decarboxylase PanD and the Unfoldase ClpC1. ACS Infect Dis 2017; 3:492-501. [PMID: 28271875 PMCID: PMC5514395 DOI: 10.1021/acsinfecdis.7b00017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Through mutant selection on agar containing pyrazinoic acid (POA), the bioactive form of the prodrug pyrazinamide (PZA), we recently showed that missense mutations in the aspartate decarboxylase PanD and the unfoldase ClpC1, and loss-of-function mutation of polyketide synthases Mas and PpsA-E involved in phthiocerol dimycocerosate synthesis, cause resistance to POA and PZA in Mycobacterium tuberculosis. Here we first asked whether these in vitro-selected POA/PZA-resistant mutants are attenuated in vivo, to potentially explain the lack of evidence of these mutations among PZA-resistant clinical isolates. Infection of mice with panD, clpC1, and mas/ppsA-E mutants showed that whereas growth of clpC1 and mas/ppsA-E mutants was attenuated, the panD mutant grew as well as the wild-type. To determine whether these resistance mechanisms can emerge within the host, mice infected with wild-type M. tuberculosis were treated with POA, and POA-resistant colonies were confirmed for PZA and POA resistance. Genome sequencing revealed that 82 and 18% of the strains contained missense mutations in panD and clpC1, respectively. Consistent with their lower fitness and POA resistance level, independent mas/ppsA-E mutants were not found. In conclusion, we show that the POA/PZA resistance mechanisms due to panD and clpC1 missense mutations are recapitulated in vivo. Whereas the representative clpC1 mutant was attenuated for growth in the mouse infection model, providing a possible explanation for their absence among clinical isolates, the growth kinetics of the representative panD mutant was unaffected. Why POA/PZA resistance-conferring panD mutations are observed in POA-treated mice but not yet among clinical strains isolated from PZA-treated patients remains to be determined.
Collapse
Affiliation(s)
- Pooja Gopal
- Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| | - Rokeya Tasneen
- Center for Tuberculosis
Research, Johns Hopkins University, Baltimore, Maryland, United States
| | - Michelle Yee
- Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| | - Jean-Philippe Lanoix
- Department of Infectious Diseases, University Hospital of Amiens-Picardie, Amiens, France
| | - Jansy Sarathy
- Public
Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - George Rasic
- Public
Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Liping Li
- Public
Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Véronique Dartois
- Public
Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Eric Nuermberger
- Center for Tuberculosis
Research, Johns Hopkins University, Baltimore, Maryland, United States
| | - Thomas Dick
- Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Republic of Singapore
| |
Collapse
|
170
|
Nusrath Unissa A, Hanna LE. Molecular mechanisms of action, resistance, detection to the first-line anti tuberculosis drugs: Rifampicin and pyrazinamide in the post whole genome sequencing era. Tuberculosis (Edinb) 2017; 105:96-107. [DOI: 10.1016/j.tube.2017.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/02/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022]
|
171
|
Njire M, Wang N, Wang B, Tan Y, Cai X, Liu Y, Mugweru J, Guo J, Hameed HMA, Tan S, Liu J, Yew WW, Nuermberger E, Lamichhane G, Liu J, Zhang T. Pyrazinoic Acid Inhibits a Bifunctional Enzyme in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2017; 61:e00070-17. [PMID: 28438933 PMCID: PMC5487608 DOI: 10.1128/aac.00070-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/01/2017] [Indexed: 11/20/2022] Open
Abstract
Pyrazinamide (PZA), an indispensable component of modern tuberculosis treatment, acts as a key sterilizing drug. While the mechanism of activation of this prodrug into pyrazinoic acid (POA) by Mycobacterium tuberculosis has been extensively studied, not all molecular determinants that confer resistance to this mysterious drug have been identified. Here, we report how a new PZA resistance determinant, the Asp67Asn substitution in Rv2783, confers M. tuberculosis resistance to PZA. Expression of the mutant allele but not the wild-type allele in M. tuberculosis recapitulates the PZA resistance observed in clinical isolates. In addition to catalyzing the metabolism of RNA and single-stranded DNA, Rv2783 also metabolized ppGpp, an important signal transducer involved in the stringent response in bacteria. All catalytic activities of the wild-type Rv2783 but not the mutant were significantly inhibited by POA. These results, which indicate that Rv2783 is a target of PZA, provide new insight into the molecular mechanism of the sterilizing activity of this drug and a basis for improving the molecular diagnosis of PZA resistance and developing evolved PZA derivatives to enhance its antituberculosis activity.
Collapse
Affiliation(s)
- Moses Njire
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- School of Life Science, University of Science and Technology of China, Hefei, China
| | - Bangxing Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yaoju Tan
- State Key Laboratory of Respiratory Disease, Department of Clinical Laboratory, Guangzhou Chest Hospital, Guangzhou, China
| | - Xingshan Cai
- State Key Laboratory of Respiratory Disease, Department of Clinical Laboratory, Guangzhou Chest Hospital, Guangzhou, China
| | - Yanwen Liu
- State Key Laboratory of Respiratory Disease, Department of Clinical Laboratory, Guangzhou Chest Hospital, Guangzhou, China
| | - Julius Mugweru
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jintao Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - H M Adnan Hameed
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shouyong Tan
- State Key Laboratory of Respiratory Disease, Department of Clinical Laboratory, Guangzhou Chest Hospital, Guangzhou, China
| | - Jianxiong Liu
- State Key Laboratory of Respiratory Disease, Department of Clinical Laboratory, Guangzhou Chest Hospital, Guangzhou, China
| | - Wing Wai Yew
- Stanley Ho Centre for Emerging Infectious Diseases, the Chinese University of Hong Kong, Hong Kong, China
| | - Eric Nuermberger
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gyanu Lamichhane
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jinsong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
172
|
Wang L, Xu M, Southall N, Zheng W, Wang S. A High-Throughput Assay for Developing Inhibitors of PhoP, a Virulence Factor of Mycobacterium tuberculosis. Comb Chem High Throughput Screen 2017; 19:855-864. [PMID: 27748178 DOI: 10.2174/1386207319666161010163249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tuberculosis (TB) kills over 1.5 million people per year despite the available anti-TB drugs. The long duration needed to treat TB by the current TB drugs, which target the essential cellular activities, inevitably leads to the emergence of drug-resistance. The emergence of drug-resistant TB prompts for an urgent need for new and more effective drugs. OBJECTIVE The response regulator PhoP, an essential virulence factor of Mycobacterium tuberculosis (MTB), is an attractive target for developing novel anti- TB drugs. This study aims to develop a robust high-throughput screening assay to identify PhoP inhibitors that disrupt the PhoP-DNA binding. METHOD Guided by the crystal structure of the PhoP-DNA complex, we designed and developed an assay based on Foster resonance energy transfer (FRET) by labeling Cy3 on the DNA and Cy5 on PhoP. We screened compound libraries for inhibitors that dissociated the PhoP-DNA complex by detection of the FRET signal. Hits were confirmed for their direct binding to PhoP by thermal shift assays. RESULTS From a test screening of ~6,000 bioactive compounds and approved drugs, three active compounds were identified that directly bound to PhoP and inhibited the PhoP-DNA interactions. These three PhoP inhibitors can be further developed to improve potency and are useful to study the mechanism of inhibition. CONCLUSION Our results demonstrated that this FRET-based PhoP-DNA binding assay is valid for additional compound library screening to identify new leads for developing novel TB drugs that target the virulence of MTB.
Collapse
Affiliation(s)
| | | | | | | | - Shuishu Wang
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd. Bethesda, MD 20814, USA.. United States
| |
Collapse
|
173
|
Ramirez-Busby SM, Rodwell TC, Fink L, Catanzaro D, Jackson RL, Pettigrove M, Catanzaro A, Valafar F. A Multinational Analysis of Mutations and Heterogeneity in PZase, RpsA, and PanD Associated with Pyrazinamide Resistance in M/XDR Mycobacterium tuberculosis. Sci Rep 2017; 7:3790. [PMID: 28630430 PMCID: PMC5476565 DOI: 10.1038/s41598-017-03452-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/28/2017] [Indexed: 11/28/2022] Open
Abstract
Pyrazinamide (PZA) is an important first-line drug in all existing and new tuberculosis (TB) treatment regimens. PZA-resistance in M. tuberculosis is increasing, especially among M/XDR cases. Noted issues with PZA Drug Susceptibility Testing (DST) have driven the search for alternative tests. This study provides a comprehensive assessment of PZA molecular diagnostics in M/XDR TB cases. A set of 296, mostly XDR, clinical M. tuberculosis isolates from four countries were subjected to DST for eight drugs, confirmatory Wayne's assay, and whole-genome sequencing. Three genes implicated in PZA resistance, pncA, rpsA, and panD were investigated. Assuming all non-synonymous mutations cause resistance, we report 90% sensitivity and 65% specificity for a pncA-based molecular test. The addition of rpsA and panD potentially provides 2% increase in sensitivity. Molecular heterogeneity in pncA was associated with resistance and should be evaluated as a diagnostic tool. Mutations near the N-terminus and C-terminus of PZase were associated with East-Asian and Euro-American lineages, respectively. Finally, Euro-American isolates are most likely to have a wild-type PZase and escape molecular detection. Overall, the 8-10% resistance without markers may point to alternative mechanisms of resistance. Confirmatory mutagenesis may improve the disconcertingly low specificity but reduce sensitivity since not all mutations may cause resistance.
Collapse
Affiliation(s)
- S M Ramirez-Busby
- Biological and Medical Informatics Research Center, San Diego State University, San Diego, California, USA
| | - T C Rodwell
- Department of Medicine, University of California, San Diego, California, USA
| | - L Fink
- Biological and Medical Informatics Research Center, San Diego State University, San Diego, California, USA
| | - D Catanzaro
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA
| | - R L Jackson
- Department of Medicine, University of California, San Diego, California, USA
| | - M Pettigrove
- Department of Medicine, University of California, San Diego, California, USA
| | - A Catanzaro
- Department of Medicine, University of California, San Diego, California, USA
| | - F Valafar
- Biological and Medical Informatics Research Center, San Diego State University, San Diego, California, USA.
| |
Collapse
|
174
|
Gao M, Gao J, Du J, Liu Y, Zhang Y, Ma L, Mi F, Li L, Tang S. Efficacy of ultra-short course chemotherapy for new smear positive drug susceptible pulmonary tuberculosis: study protocol of a multicenter randomized controlled clinical trial. BMC Infect Dis 2017. [PMID: 28629333 PMCID: PMC5474865 DOI: 10.1186/s12879-017-2505-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Shortening the standard 6-month treatment for drug-susceptible pulmonary tuberculosis (DS-PTB) would be a major improvement for TB case management and disease control. Methods We are conducting a randomized, open-label, controlled, non-inferiority trial involving patients with smear-positive, newly diagnosed DS-PTB cases nationwide to assess the efficacy and safety of two 4.5- month regimens in comparison to the standard 6-month WHO recommended regimen. The regimen used in one experiment group is a 4.5-month fluoroquinolone-containing regimen, which consists of full course of levofloxacin, isoniazid (H), rifampin (R), parazinamid (Z) and ethambutol (E). Regimen used in the second experiment group includes 4.5-month full course of H, R, Z, E with levofloxacin removed. Patients in the control group, receive H, R, Z and E for 2 months, followed by 4 months of H and R. The primary endpoint is treatment failure or relapse within 24 month after treatment completion. Discussion Results from this trial along with other studies will contribute to the science of constructing a shorter, effective and safe regiment for TB patients. Trial registration The protocol has been registered on ClinicalTrials.gov on 2 September,2016 with identifier NCT02901288.
Collapse
Affiliation(s)
- Mengqiu Gao
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Jingtao Gao
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Jian Du
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Yuhong Liu
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Yao Zhang
- Family Health International 360 (FHI360), Beijing, 100020, China
| | - Liping Ma
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Fengling Mi
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China
| | - Liang Li
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China.
| | - Shenjie Tang
- Beijing Chest Hospital, Capital Medical University, Tuberculosis and Thoracic Tumor Research Institute, Beijing101149, Beijing, China.
| | | |
Collapse
|
175
|
Sun X, Li Y, He W, Ji C, Xia P, Wang Y, Du S, Li H, Raikhel N, Xiao J, Guo H. Pyrazinamide and derivatives block ethylene biosynthesis by inhibiting ACC oxidase. Nat Commun 2017; 8:15758. [PMID: 28604689 PMCID: PMC5472784 DOI: 10.1038/ncomms15758] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 04/25/2017] [Indexed: 12/30/2022] Open
Abstract
Ethylene is an important phytohormone that promotes the ripening of fruits and senescence of flowers thereby reducing their shelf lives. Specific ethylene biosynthesis inhibitors would help to decrease postharvest loss. Here, we identify pyrazinamide (PZA), a clinical drug used to treat tuberculosis, as an inhibitor of ethylene biosynthesis in Arabidopsis thaliana, using a chemical genetics approach. PZA is converted to pyrazinecarboxylic acid (POA) in plant cells, suppressing the activity of 1-aminocyclopropane-1-carboxylic acid oxidase (ACO), the enzyme catalysing the final step of ethylene formation. The crystal structures of Arabidopsis ACO2 in complex with POA or 2-Picolinic Acid (2-PA), a POA-related compound, reveal that POA/2-PA bind at the active site of ACO, preventing the enzyme from interacting with its natural substrates. Our work suggests that PZA and its derivatives may be promising regulators of plant metabolism, in particular ethylene biosynthesis.
Collapse
Affiliation(s)
- Xiangzhong Sun
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Beijing 100871, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yaxin Li
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Wenrong He
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China.,Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, California 92507, USA
| | - Chenggong Ji
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Peixue Xia
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Yichuan Wang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Shuo Du
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Hongjiang Li
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China.,Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, California 92507, USA
| | - Natasha Raikhel
- Center for Plant Cell Biology, Department of Botany and Plant Sciences, University of California, Riverside, California 92507, USA
| | - Junyu Xiao
- The State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Hongwei Guo
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
176
|
Nasiri MJ, Haeili M, Ghazi M, Goudarzi H, Pormohammad A, Imani Fooladi AA, Feizabadi MM. New Insights in to the Intrinsic and Acquired Drug Resistance Mechanisms in Mycobacteria. Front Microbiol 2017; 8:681. [PMID: 28487675 PMCID: PMC5403904 DOI: 10.3389/fmicb.2017.00681] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/04/2017] [Indexed: 01/25/2023] Open
Abstract
Infectious diseases caused by clinically important Mycobacteria continue to be an important public health problem worldwide primarily due to emergence of drug resistance crisis. In recent years, the control of tuberculosis (TB), the disease caused by Mycobacterium tuberculosis (MTB), is hampered by the emergence of multidrug resistance (MDR), defined as resistance to at least isoniazid (INH) and rifampicin (RIF), two key drugs in the treatment of the disease. Despite the availability of curative anti-TB therapy, inappropriate and inadequate treatment has allowed MTB to acquire resistance to the most important anti-TB drugs. Likewise, for most mycobacteria other than MTB, the outcome of drug treatment is poor and is likely related to the high levels of antibiotic resistance. Thus, a better knowledge of the underlying mechanisms of drug resistance in mycobacteria could aid not only to select the best therapeutic options but also to develop novel drugs that can overwhelm the existing resistance mechanisms. In this article, we review the distinctive mechanisms of antibiotic resistance in mycobacteria.
Collapse
Affiliation(s)
- Mohammad J. Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical SciencesTehran, Iran
| | - Mehri Haeili
- Department of Biology, Faculty of Natural Sciences, University of TabrizTabriz, Iran
| | - Mona Ghazi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical SciencesTehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical SciencesTehran, Iran
| | - Ali Pormohammad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical SciencesTehran, Iran
| | - Abbas A. Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical SciencesTehran, Iran
| | - Mohammad M. Feizabadi
- Department of Microbiology, School of Medicine, Tehran University of Medical SciencesTehran, Iran
- Thoracic Research Center, Imam Khomeini Hospital, Tehran University of Medical SciencesTehran, Iran
| |
Collapse
|
177
|
Hong WD, Gibbons PD, Leung SC, Amewu R, Stocks PA, Stachulski A, Horta P, Cristiano MLS, Shone AE, Moss D, Ardrey A, Sharma R, Warman AJ, Bedingfield PTP, Fisher NE, Aljayyoussi G, Mead S, Caws M, Berry NG, Ward SA, Biagini GA, O'Neill PM, Nixon GL. Rational Design, Synthesis, and Biological Evaluation of Heterocyclic Quinolones Targeting the Respiratory Chain of Mycobacterium tuberculosis. J Med Chem 2017; 60:3703-3726. [PMID: 28304162 DOI: 10.1021/acs.jmedchem.6b01718] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A high-throughput screen (HTS) was undertaken against the respiratory chain dehydrogenase component, NADH:menaquinone oxidoreductase (Ndh) of Mycobacterium tuberculosis (Mtb). The 11000 compounds were selected for the HTS based on the known phenothiazine Ndh inhibitors, trifluoperazine and thioridazine. Combined HTS (11000 compounds) and in-house screening of a limited number of quinolones (50 compounds) identified ∼100 hits and four distinct chemotypes, the most promising of which contained the quinolone core. Subsequent Mtb screening of the complete in-house quinolone library (350 compounds) identified a further ∼90 hits across three quinolone subtemplates. Quinolones containing the amine-based side chain were selected as the pharmacophore for further modification, resulting in metabolically stable quinolones effective against multi drug resistant (MDR) Mtb. The lead compound, 42a (MTC420), displays acceptable antituberculosis activity (Mtb IC50 = 525 nM, Mtb Wayne IC50 = 76 nM, and MDR Mtb patient isolates IC50 = 140 nM) and favorable pharmacokinetic and toxicological profiles.
Collapse
Affiliation(s)
- W David Hong
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Peter D Gibbons
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Suet C Leung
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Richard Amewu
- Department of Chemistry, University of Ghana , P.O. Box LG56, Legon-Accra, Ghana
| | - Paul A Stocks
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Andrew Stachulski
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Pedro Horta
- CCMAR and Department of Chemistry and Pharmacy, University of Algarve , 8005-139 Faro, Portugal
| | - Maria L S Cristiano
- CCMAR and Department of Chemistry and Pharmacy, University of Algarve , 8005-139 Faro, Portugal
| | - Alison E Shone
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Darren Moss
- School of Pharmacy, Keele University , Keele ST5 5BG, U.K
| | - Alison Ardrey
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Raman Sharma
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Ashley J Warman
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Paul T P Bedingfield
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Nicholas E Fisher
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Ghaith Aljayyoussi
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Sally Mead
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Maxine Caws
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Neil G Berry
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Stephen A Ward
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Giancarlo A Biagini
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine , Pembroke Place, Liverpool L3 5QA, U.K
| | - Paul M O'Neill
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| | - Gemma L Nixon
- Department of Chemistry, University of Liverpool , Liverpool L69 7ZD, U.K
| |
Collapse
|
178
|
Fan Y, Dai Y, Hou M, Wang H, Yao H, Guo C, Lin D, Liao X. Structural basis for ribosome protein S1 interaction with RNA in trans-translation of Mycobacterium tuberculosis. Biochem Biophys Res Commun 2017; 487:268-273. [PMID: 28412369 DOI: 10.1016/j.bbrc.2017.04.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/11/2017] [Indexed: 11/24/2022]
Abstract
Ribosomal protein S1 (RpsA), the largest 30S protein in ribosome, plays a significant role in translation and trans-translation. In Mycobacterium tuberculosis, the C-terminus of RpsA is known as tuberculosis drug target of pyrazinoic acid, which inhibits the interaction between MtRpsA and tmRNA in trans-translation. However, the molecular mechanism underlying the interaction of MtRpsA with tmRNA remains unknown. We herein analyzed the interaction of the C-terminal domain of MtRpsA with three RNA fragments poly(A), sMLD and pre-sMLD. NMR titration analysis revealed that the RNA binding sites on MtRpsACTD are mainly located in the β2, β3 and β5 strands and the adjacent L3 loop of the S1 domain. Fluorescence experiments determined the MtRpsACTD binding to RNAs are in the micromolar affinity range. Sequence analysis also revealed conserved residues in the mapped RNA binding region. Residues L304, V305, G308, F310, H322, I323, R357 and I358 were verified to be the key residues influencing the interaction between MtRpsACTD and pre-sMLD. Molecular docking further confirmed that the poly(A)-like sequence and sMLD of tmRNA are all involved in the protein-RNA interaction, through charged interaction and hydrogen bonds. The results will be beneficial for designing new anti-tuberculosis drugs.
Collapse
Affiliation(s)
- Yi Fan
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yazhuang Dai
- School of Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Meijing Hou
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Huilin Wang
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongwei Yao
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chenyun Guo
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Xinli Liao
- Key Laboratory of Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
179
|
Non- pncA Gene-Mutated but Pyrazinamide-Resistant Mycobacterium tuberculosis: Why Is That? J Clin Microbiol 2017; 55:1920-1927. [PMID: 28404681 DOI: 10.1128/jcm.02532-16] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/02/2017] [Indexed: 01/08/2023] Open
Abstract
Pyrazinamide (PZA) is a key component for the effective treatment of drug-susceptible and PZA-susceptible multidrug-resistant (MDRPZA-S) tuberculosis (TB). pncA gene mutations are usually detected in a clear majority (>90%) of PZA-resistant strains but obviously not in all. Rapid and reliable PZA drug susceptibility testing (DST) is critical whenever PZA is to be used in a treatment regimen, not least for the treatment of MDRPZA-S TB. In this study, we selected 26 PZA-resistant isolates reported to carry a wild-type pncA gene. To confirm resistance, susceptibility testing was repeated using 100 mg/liter and 200 mg/liter PZA for all the 26 isolates and Sanger sequencing was repeated on the 18 isolates that remained PZA resistant. Apart from the eight isolates initially misclassified as PZA resistant, the retests identified three factors responsible for the phenotype-genotype discrepancy: panD or rpsA mutations identified by whole-genome sequencing (WGS) (n = 7), heteroresistance (n = 8), and mixed populations with Mycobacterium avium (n = 3). Additionally, we performed WGS on 400 PZA-susceptible isolates and 15 consecutive MDRPZA-R clinical isolates. Of the 400 PZA-susceptible isolates, only 1 harbored a nonsynonymous pncA mutation (Thr87Met), whereas a nonsynonymous rpsA mutation was found in 17 isolates. None of these isolates carried a nonsynonymous panD mutation, while all 15 of the MDRPZA-R isolates harbored a nonsynonymous pncA mutation. Our findings indicate that it is necessary to consider the occurrence of panD mutations in PZA-resistant isolates, as well as heteroresistance, for the development and evaluation of new molecular techniques to ensure high-quality DST performance. The identification of nonsynonymous rpsA mutations in both PZA-susceptible and PZA-resistant isolates also implies that further studies are needed in order to determine the role of rpsA in PZA resistance.
Collapse
|
180
|
Identification of Mycobacterial RplJ/L10 and RpsA/S1 Proteins as Novel Targets for CD4 + T Cells. Infect Immun 2017; 85:IAI.01023-16. [PMID: 28115505 PMCID: PMC5364311 DOI: 10.1128/iai.01023-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Tuberculosis (TB) due to Mycobacterium tuberculosis remains a major global infectious disease problem, and a more efficacious vaccine is urgently needed for the control and prevention of disease caused by this organism. We previously reported that a genetically modified strain of Mycobacterium smegmatis called IKEPLUS is a promising TB vaccine candidate. Since protective immunity induced by IKEPLUS is dependent on antigen-specific CD4+ T cell memory, we hypothesized that the specificity of the CD4+ T cell response was a critical feature of this protection. Using in vitro assays of interferon gamma production (enzyme-linked immunosorbent spot [ELISPOT] assays) by splenocytes from IKEPLUS-immunized C57BL/6J mice, we identified an immunogenic peptide within the mycobacterial ribosomal large subunit protein RplJ, encoded by the Rv0651 gene. In a complementary approach, we generated major histocompatibility complex (MHC) class II-restricted T cell hybridomas from IKEPLUS-immunized mice. Screening of these T cell hybridomas against IKEPLUS and ribosomes enriched from IKEPLUS suggested that the CD4+ T cell response in IKEPLUS-immunized mice was dominated by the recognition of multiple components of the mycobacterial ribosome. Importantly, CD4+ T cells specific for mycobacterial ribosomes accumulate to significant levels in the lungs of IKEPLUS-immunized mice following aerosol challenge with virulent M. tuberculosis, consistent with a role for these T cells in protective host immunity in TB. The identification of CD4+ T cell responses to defined ribosomal protein epitopes expands the range of antigenic targets for adaptive immune responses to M. tuberculosis and may help to inform the design of more effective vaccines against tuberculosis.
Collapse
|
181
|
Jung IP, Ha NR, Kim AR, Kim SH, Yoon MY. Mutation analysis of the interactions between Mycobacterium tuberculosis caseinolytic protease C1 (ClpC1) and ecumicin. Int J Biol Macromol 2017; 101:348-357. [PMID: 28342755 DOI: 10.1016/j.ijbiomac.2017.03.126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/21/2017] [Accepted: 03/21/2017] [Indexed: 10/19/2022]
Abstract
Ecumicin is a well-known and potent inhibitor of Mycobacterium tuberculosis. Although the target of ecumicin is caseinolytic protease C1 (ClpC1), the exact mechanism by which ecumicin inhibits ClpC1 has not been identified. To analyze ecumicin's action on ClpC1, site-directed mutagenesis was performed on its binding site. The estimated binding residues within ClpC1 to ecumicin were selected via in silico analysis using molecular docking. The selected residues were mutated by site-directed mutagenesis and the effects on ecumicin binding were analyzed. Mutation at the R83 residue, especially the R83A mutation, in ClpC1 resulted in strong resistance to ATPase activation and inhibition of proteolytic activity. In addition, binding of ecumicin to the R83A ClpC1 N-terminal domain (residues 1-145) was not observed in native gel analysis. These results reveal that the R83 residue plays an important role in the binding of ecumicin. This result provides a basis for the development of an anti-tuberculosis agent based on ecumicin derivatives.
Collapse
Affiliation(s)
- In-Pil Jung
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Na-Reum Ha
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - A-Ru Kim
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Sang-Heon Kim
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Moon-Young Yoon
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
182
|
3-Substituted N-Benzylpyrazine-2-carboxamide Derivatives: Synthesis, Antimycobacterial and Antibacterial Evaluation. Molecules 2017; 22:molecules22030495. [PMID: 28335571 PMCID: PMC6155222 DOI: 10.3390/molecules22030495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/06/2017] [Accepted: 03/17/2017] [Indexed: 11/17/2022] Open
Abstract
A series of substituted N-benzyl-3-chloropyrazine-2-carboxamides were prepared as positional isomers of 5-chloro and 6-chloro derivatives, prepared previously. During the aminolysis of the acyl chloride, the simultaneous substitution of chlorine with benzylamino moiety gave rise to N-benzyl-3-(benzylamino)pyrazine-2-carboxamides as side products, in some cases. Although not initially planned, the reaction conditions were modified to populate this double substituted series. The final compounds were tested against four mycobacterial strains. N-(2-methylbenzyl)-3-((2-methylbenzyl)amino)pyrazine-2-carboxamide (1a) and N-(3,4-dichlorobenzyl)-3-((3,4-dichlorobenzyl)amino)pyrazine-2-carboxamide (9a) proved to be the most effective against Mycobacterium tuberculosis H37Rv, with MIC = 12.5 μg·mL-1. Compounds were screened for antibacterial activity. The most active compound was 3-chloro-N-(2-chlorobenzyl)pyrazine-2-carboxamide (5) against Staphylococcus aureus with MIC = 7.81 μM, and Staphylococcus epidermidis with MIC = 15.62 μM. HepG2 in vitro cytotoxicity was evaluated for the most active compounds; however, no significant toxicity was detected. Compound 9a was docked to several conformations of the enoyl-ACP-reductase of Mycobacterium tuberculosis. In some cases, it was capable of H-bond interactions, typical for most of the known inhibitors.
Collapse
|
183
|
Khadem-Maaref M, Mehrnejad F, Phirouznia A. Effects of metal-ion replacement on pyrazinamidase activity: A quantum mechanical study. J Mol Graph Model 2017; 73:24-29. [PMID: 28214629 DOI: 10.1016/j.jmgm.2017.01.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/27/2017] [Accepted: 01/30/2017] [Indexed: 11/26/2022]
Abstract
Pyrazinamidase (PZase), a metalloenzyme, is responsible for acidic modification of pyrazinamide (PZA), a drug used in tuberculosis treatment. The metal coordination site of the enzyme is able to coordinate various divalent metal cofactors. Previous experimental studies have demonstrated that metal ions, such as Co2+, Mn2+, and Zn2+, are able to reactivate metal-depleted PZase, while others including Cu2+, Fe2+, and Mg2+, cannot restore activity. In this study, we investigated binding of various metal ions to the metal coordination site (MCS) of the enzyme using quantum mechanical calculations. We calculated the metal-ligand (residue) binding energy and the atomic partial charges in the presence of various ions. The results indicated that the tendency of alkaline earth metals to bind to PZase MCS is very low and not suitable for enzyme structural and catalytic function. In contrast, Co2+ and Ni2+ ions have very high binding affinity and are favorable to the structural and functional properties of the enzyme. Furthermore, we observed that the rate at which Ni2+, Co2+ and Fe2+ ions in PZase MCS polarize the OH bond of coordinated water molecules is much higher than the polarization rate created by other ions. This finding suggests that the coordination of Ni2+, Co2+, or Fe2+ to PZase facilitates the deprotonation of coordinated water molecules to generate a nucleophile that catalyzes the enzymatic reaction.
Collapse
Affiliation(s)
- Mahmoud Khadem-Maaref
- Department of Physics, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Faramarz Mehrnejad
- Department of Life Sciences Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
| | - Arash Phirouznia
- Department of Physics, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran.
| |
Collapse
|
184
|
Van den Bergh B, Fauvart M, Michiels J. Formation, physiology, ecology, evolution and clinical importance of bacterial persisters. FEMS Microbiol Rev 2017; 41:219-251. [DOI: 10.1093/femsre/fux001] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
|
185
|
Zhang S, Chen J, Shi W, Cui P, Zhang J, Cho S, Zhang W, Zhang Y. Mutation in clpC1 encoding an ATP-dependent ATPase involved in protein degradation is associated with pyrazinamide resistance in Mycobacterium tuberculosis. Emerg Microbes Infect 2017; 6:e8. [PMID: 28196969 PMCID: PMC5322326 DOI: 10.1038/emi.2017.1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Shuo Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
- Shandong Medicinal Biotechnology Centre, Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Jiazhen Chen
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wanliang Shi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Peng Cui
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jia Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Sanghyun Cho
- College of Pharmacy, Institute for Tuberculosis Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wenhong Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
186
|
Synthesis of Novel Pyrazinamide Derivatives Based on 3-Chloropyrazine-2-carboxamide and Their Antimicrobial Evaluation. Molecules 2017; 22:molecules22020223. [PMID: 28157178 PMCID: PMC6155776 DOI: 10.3390/molecules22020223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/27/2017] [Indexed: 12/19/2022] Open
Abstract
Aminodehalogenation of 3-chloropyrazine-2-carboxamide with variously substituted benzylamines yielded a series of fifteen 3-benzylaminopyrazine-2-carboxamides. Four compounds possessed in vitro whole cell activity against Mycobacterium tuberculosis H37Rv that was at least equivalent to that of the standard pyrazinamide. MIC values ranged from 6 to 42 µM. The best MIC (6 µM) was displayed by 3-[(4-methylbenzyl)amino]pyrazine-2-carboxamide (8) that also showed low cytotoxicity in the HepG2 cell line (IC50 ≥ 250 µM). Only moderate activity against Enterococcus faecalis and Staphylococcus aureus was observed. No activity was detected against any of tested fungal strains. Molecular docking with mycobacterial enoyl-ACP reductase (InhA) was performed to investigate the possible target of the prepared compounds. Active compounds shared common binding interactions of known InhA inhibitors. Antimycobacterial activity of the title compounds was compared to the previously published benzylamino-substituted pyrazines with differing substitution on the pyrazine core (carbonitrile moiety). The title series possessed comparable activity and lower cytotoxicity than molecules containing a carbonitrile group on the pyrazine ring.
Collapse
|
187
|
Zhang Z, Ordonez AA, Smith-Jones P, Wang H, Gogarty KR, Daryaee F, Bambarger LE, Chang YS, Jain SK, Tonge PJ. The biodistribution of 5-[18F]fluoropyrazinamide in Mycobacterium tuberculosis-infected mice determined by positron emission tomography. PLoS One 2017; 12:e0170871. [PMID: 28151985 PMCID: PMC5289470 DOI: 10.1371/journal.pone.0170871] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/11/2017] [Indexed: 02/03/2023] Open
Abstract
5-[18F]F-pyrazinamide (5-[18F]F-PZA), a radiotracer analog of the first-line tuberculosis drug pyrazinamide (PZA), was employed to determine the biodistribution of PZA using PET imaging and ex vivo analysis. 5-[18F]F-PZA was synthesized in 60 min using a halide exchange reaction. The overall decay-corrected yield of the reaction was 25% and average specific activity was 2.6 × 106 kBq (70 mCi)/μmol. The biodistribution of 5-[18F]F-PZA was examined in a pulmonary Mycobacterium tuberculosis mouse model, where rapid distribution of the tracer to the lung, heart, liver, kidney, muscle, and brain was observed. The concentration of 5-[18F]F-PZA was not significantly different between infected and uninfected lung tissue. Biochemical and microbiological studies revealed substantial differences between 5-F-PZA and PZA. 5-F-PZA was not a substrate for pyrazinamidase, the bacterial enzyme that activates PZA, and the minimum inhibitory concentration for 5-F-PZA against M. tuberculosis was more than 100-fold higher than that for PZA.
Collapse
Affiliation(s)
- Zhuo Zhang
- Institute for Chemical Biology & Drug Discovery, Department of Chemistry and Department of Radiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research, Center for Tuberculosis Research and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Peter Smith-Jones
- The Facility for Experimental Radiopharmaceutical Manufacturing, Department of Psychiatry, Stony Brook University, Stony Brook, New York, United States of America
| | - Hui Wang
- Institute for Chemical Biology & Drug Discovery, Department of Chemistry and Department of Radiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Kayla R. Gogarty
- Institute for Chemical Biology & Drug Discovery, Department of Chemistry and Department of Radiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Fereidoon Daryaee
- Institute for Chemical Biology & Drug Discovery, Department of Chemistry and Department of Radiology, Stony Brook University, Stony Brook, New York, United States of America
| | - Lauren E. Bambarger
- Center for Infection and Inflammation Imaging Research, Center for Tuberculosis Research and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yong S. Chang
- Center for Infection and Inflammation Imaging Research, Center for Tuberculosis Research and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Center for Tuberculosis Research and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail: (SKJ); (PJT)
| | - Peter J. Tonge
- Institute for Chemical Biology & Drug Discovery, Department of Chemistry and Department of Radiology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail: (SKJ); (PJT)
| |
Collapse
|
188
|
Missense Mutations in the Unfoldase ClpC1 of the Caseinolytic Protease Complex Are Associated with Pyrazinamide Resistance in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2017; 61:AAC.02342-16. [PMID: 27872068 PMCID: PMC5278685 DOI: 10.1128/aac.02342-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/12/2016] [Indexed: 11/20/2022] Open
Abstract
Previously, we showed that mutations in Mycobacterium tuberculosispanD, involved in coenzyme A biosynthesis, cause resistance against pyrazinoic acid, the bioactive component of the prodrug pyrazinamide. To identify additional resistance mechanisms, we isolated mutants resistant against pyrazinoic acid and subjected panD wild-type strains to whole-genome sequencing. Eight of the nine resistant strains harbored missense mutations in the unfoldase ClpC1 associated with the caseinolytic protease complex.
Collapse
|
189
|
Long-Chain Fatty Acyl Coenzyme A Ligase FadD2 Mediates Intrinsic Pyrazinamide Resistance in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2017; 61:AAC.02130-16. [PMID: 27855077 DOI: 10.1128/aac.02130-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/29/2016] [Indexed: 11/20/2022] Open
Abstract
Pyrazinamide (PZA) is a first-line tuberculosis (TB) drug that has been in clinical use for 60 years yet still has an unresolved mechanism of action. Based upon the observation that the minimum concentration of PZA required to inhibit the growth of Mycobacterium tuberculosis is approximately 1,000-fold higher than that of other first-line drugs, we hypothesized that M. tuberculosis expresses factors that mediate intrinsic resistance to PZA. To identify genes associated with intrinsic PZA resistance, a library of transposon-mutagenized Mycobacterium bovis BCG strains was screened for strains showing hypersusceptibility to the active form of PZA, pyrazinoic acid (POA). Disruption of the long-chain fatty acyl coenzyme A (CoA) ligase FadD2 enhanced POA susceptibility by 16-fold on agar medium, and the wild-type level of susceptibility was restored upon expression of fadD2 from an integrating mycobacterial vector. Consistent with the recent observation that POA perturbs mycobacterial CoA metabolism, the fadD2 mutant strain was more vulnerable to POA-mediated CoA depletion than the wild-type strain. Ectopic expression of the M. tuberculosis pyrazinamidase PncA, necessary for conversion of PZA to POA, in the fadD2 transposon insertion mutant conferred at least a 16-fold increase in PZA susceptibility under active growth conditions in liquid culture at neutral pH. Importantly, deletion of fadD2 in M. tuberculosis strain H37Rv also resulted in enhanced susceptibility to POA. These results indicate that FadD2 is associated with intrinsic PZA and POA resistance and provide a proof of concept for the target-based potentiation of PZA activity in M. tuberculosis.
Collapse
|
190
|
Guo HL, Hassan HM, Ding PP, Wang SJ, Chen X, Wang T, Sun LX, Zhang LY, Jiang ZZ. Pyrazinamide-induced hepatotoxicity is alleviated by 4-PBA via inhibition of the PERK-eIF2α-ATF4-CHOP pathway. Toxicology 2017; 378:65-75. [PMID: 28063906 DOI: 10.1016/j.tox.2017.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/11/2016] [Accepted: 01/03/2017] [Indexed: 12/30/2022]
Abstract
Pyrazinamide (PZA)-induced serious liver injury, but the exact mechanism of PZA-induces hepatotoxicity remains controversial. Endoplasmic reticulum (ER) stress-caused cell apoptosis plays a critical role in the development of drug-induced liver injury (DILI). However, the direct connection between PZA toxicity and ER stress is unknown. In this study, we describe the role of ER stress in PZA induced hepatotoxicity in vivo and in vitro. We found that PZA induces apoptosis in HepG2 cells, and causes liver damage in rats, characterized by increased serum ALT, AST and TBA levels. PZA impairs antioxidant defenses, although this effect did not play an important role in resulting liver injury. The ER stress related proteins GRP78, p-PERK, p-eIF2α, ATF4, CHOP and caspase12 were activated after PZA exposure both in vivo and in vitro. Furthermore, as an ER stress inhibitor, sodium 4-phenylbutyrate (4-PBA) could ameliorate PZA toxicity in HepG2 cells and rat liver. These results have potential implications for the pathogenesis of PZA-induced hepatotoxicity in which ER stress especially PERK-eIF2α-ATF4-CHOP pathway participates in hepatocellular injury.
Collapse
Affiliation(s)
- Hong-Li Guo
- Children's Hospital of Nanjing Medical University, Nanjing, 210008, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Hozeifa M Hassan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, Faculty of Pharmacy, University of Gezira, Wad-Medani, Sudan
| | - Ping-Ping Ding
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Shao-Jie Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Xi Chen
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China
| | - Li-Xin Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
191
|
Gold B, Nathan C. Targeting Phenotypically Tolerant Mycobacterium tuberculosis. Microbiol Spectr 2017; 5:10.1128/microbiolspec.TBTB2-0031-2016. [PMID: 28233509 PMCID: PMC5367488 DOI: 10.1128/microbiolspec.tbtb2-0031-2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Indexed: 01/08/2023] Open
Abstract
While the immune system is credited with averting tuberculosis in billions of individuals exposed to Mycobacterium tuberculosis, the immune system is also culpable for tempering the ability of antibiotics to deliver swift and durable cure of disease. In individuals afflicted with tuberculosis, host immunity produces diverse microenvironmental niches that support suboptimal growth, or complete growth arrest, of M. tuberculosis. The physiological state of nonreplication in bacteria is associated with phenotypic drug tolerance. Many of these host microenvironments, when modeled in vitro by carbon starvation, complete nutrient starvation, stationary phase, acidic pH, reactive nitrogen intermediates, hypoxia, biofilms, and withholding streptomycin from the streptomycin-addicted strain SS18b, render M. tuberculosis profoundly tolerant to many of the antibiotics that are given to tuberculosis patients in clinical settings. Targeting nonreplicating persisters is anticipated to reduce the duration of antibiotic treatment and rate of posttreatment relapse. Some promising drugs to treat tuberculosis, such as rifampin and bedaquiline, only kill nonreplicating M. tuberculosisin vitro at concentrations far greater than their minimal inhibitory concentrations against replicating bacilli. There is an urgent demand to identify which of the currently used antibiotics, and which of the molecules in academic and corporate screening collections, have potent bactericidal action on nonreplicating M. tuberculosis. With this goal, we review methods of high-throughput screening to target nonreplicating M. tuberculosis and methods to progress candidate molecules. A classification based on structures and putative targets of molecules that have been reported to kill nonreplicating M. tuberculosis revealed a rich diversity in pharmacophores.
Collapse
Affiliation(s)
- Ben Gold
- Department of Microbiology & Immunology, Weill Cornell Medical College, New York, NY, 10065
| | - Carl Nathan
- Department of Microbiology & Immunology, Weill Cornell Medical College, New York, NY, 10065
| |
Collapse
|
192
|
Colameco S, Elliot MA. Non-coding RNAs as antibiotic targets. Biochem Pharmacol 2016; 133:29-42. [PMID: 28012959 DOI: 10.1016/j.bcp.2016.12.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Antibiotics inhibit a wide range of essential processes in the bacterial cell, including replication, transcription, translation and cell wall synthesis. In many instances, these antibiotics exert their effects through association with non-coding RNAs. This review highlights many classical antibiotic targets (e.g. rRNAs and the ribosome), explores a number of emerging targets (e.g. tRNAs, RNase P, riboswitches and small RNAs), and discusses the future directions and challenges associated with non-coding RNAs as antibiotic targets.
Collapse
Affiliation(s)
- Savannah Colameco
- Department of Biology and Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Marie A Elliot
- Department of Biology and Institute for Infectious Disease Research, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
193
|
Abstract
Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, uses various tactics to resist on antibiotics and evade host immunity. To control tuberculosis, antibiotics with novel mechanisms of action are urgently needed. Emerging new antibiotics and underlying novel drug targets are summarized in this paper.
Collapse
Affiliation(s)
- Nzungize Lambert
- a Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University , Chongqing , China
| | - Abualgasim Elgaili Abdalla
- a Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University , Chongqing , China.,b Department of Clinical Microbiology, College of Medical Laboratory Sciences, Omdurman , Islamic University , Omdurman , Sudan
| | - Xiangke Duan
- a Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University , Chongqing , China
| | - Jianping Xie
- a Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University , Chongqing , China
| |
Collapse
|
194
|
Qian H, Zhu K, Lu H, Lavoie M, Chen S, Zhou Z, Deng Z, Chen J, Fu Z. Contrasting silver nanoparticle toxicity and detoxification strategies in Microcystis aeruginosa and Chlorella vulgaris: New insights from proteomic and physiological analyses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2016; 572:1213-1221. [PMID: 27522289 DOI: 10.1016/j.scitotenv.2016.08.039] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 06/06/2023]
Abstract
Several studies have shown that AgNPs can be toxic to phytoplankton, but the underlying cellular mechanisms still remain largely unknown. Here we studied the toxicity and detoxification of AgNPs (and ionic silver released by the AgNPs) in a prokaryotic (Microcystis aeruginosa) and a eukaryotic (Chlorella vulgaris) freshwater phytoplankton species using a combination of proteomic, gene transcription, and physiological analyses. We show that AgNPs were more toxic to the growth, photosynthesis, antioxidant systems, and carbohydrate metabolism of M. aeruginosa than of C. vulgaris. C. vulgaris could detoxify efficiently AgNPs-induced ROS species via induction of antioxidant enzymes (superoxide dismutase or SOD, peroxidase or POD, catalase or CAT, and glutamine synthetase), allowing photosynthesis to continue unabated at growth-inhibitory AgNPs concentration. By contrast, the transcription and expression of SOD and POD in M. aeruginosa was inhibited by the same AgNPs exposure. The present study shed new lights on the AgNPs toxicity mechanisms and detoxification strategies in two freshwater algae of contrasting AgNPs sensitivity.
Collapse
Affiliation(s)
- Haifeng Qian
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Kun Zhu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haiping Lu
- College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, PR China
| | - Michel Lavoie
- Quebec-Ocean and Takuvik Joint International Research Unit, Université Laval, Québec, Canada
| | - Si Chen
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zhongjing Zhou
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Virology and Biotechnology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Zhiping Deng
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Virology and Biotechnology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Jun Chen
- College of Environment, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China.
| |
Collapse
|
195
|
Zheng X, Ning Z, Drobniewski F, Yang J, Li Q, Zhang Z, Hu Y. pncA mutations are associated with slower sputum conversion during standard treatment of multidrug-resistant tuberculosis. Int J Antimicrob Agents 2016; 49:183-188. [PMID: 28012685 DOI: 10.1016/j.ijantimicag.2016.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 09/24/2016] [Accepted: 10/02/2016] [Indexed: 11/17/2022]
Abstract
Despite the strong association between drug resistance and genetic mutations, the value of molecular diagnosis of drug resistance to guide the treatment of multidrug-resistant tuberculosis (MDR-TB) remains unclear. This is particularly relevant in resource-limited areas where it is difficult to perform drug susceptibility testing (DST). Here we investigated the association between drug susceptibility phenotypes and genotypes and treatment outcomes in patients with MDR-TB. This study enrolled 74 consecutive patients with confirmed MDR-TB between 2010 and 2011, and outcomes were followed-up over the 24-month treatment course. All of the isolates were tested for phenotypic susceptibility to second-line drugs using the Mycobacteria Growth Indicator Tube (MGIT)-based system, and genotypic mutations were assessed by DNA sequencing. Among the 74 MDR-TB isolates, 29 (39.2%) were resistant to fluoroquinolones and/or second-line injectable drugs, of which 21 (72.4%) harboured a mutation in drug resistance-related genes (gyrA, rrs or eis). In addition, 32 individuals (43.2%) also had pyrazinamide (PZA)-resistant isolates, with 28 (87.5%) containing the pncA mutation. By backward selection in the multivariate logistic regression and Cox proportional hazard models, PZA resistance and its related pncA gene mutation demonstrated a correlation with a lower likelihood of culture conversion at 8 weeks and treatment success. Meanwhile, the fluoroquinolone resistance-related gyrA gene mutation was negatively correlated with treatment success. DST for PZA and fluoroquinolones together with genetic information appears to provide a clinically useful indicator of the treatment outcome of MDR-TB in China.
Collapse
Affiliation(s)
- Xubin Zheng
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Public Health Safety (Fudan University), China
| | - Zhu Ning
- Zigong City Center for Disease Control and Prevention, Zigong City, Sichuan, China
| | | | - Jingyong Yang
- Shanghai integrated traditional Chinese and Western Medicine Hospital, Shanghai, China
| | - Qun Li
- Zigong City Center for Disease Control and Prevention, Zigong City, Sichuan, China
| | - Zhengdong Zhang
- Zigong City Center for Disease Control and Prevention, Zigong City, Sichuan, China
| | - Yi Hu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China; Ministry of Education, Key Laboratory of Public Health Safety (Fudan University), China.
| |
Collapse
|
196
|
da Silva PB, Campos DL, Ribeiro CM, da Silva IC, Pavan FR. New antimycobacterial agents in the pre-clinical phase or beyond: recent advances in patent literature (2001-2016). Expert Opin Ther Pat 2016; 27:269-282. [PMID: 27796146 DOI: 10.1080/13543776.2017.1253681] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Tuberculosis, an infectious disease, has caused more deaths worldwide than any other single infectious disease, killing more than 1.5 million people each year; equating to 4,100 deaths a day. In the past 60 years, no new drugs have been added to the first line regimen, in spite of the fact that thousands of papers have been published on drugs against tuberculosis and hundreds of drugs have received patents as new potential products. Thus, there is undoubtedly an urgent need for the deployment of new effective drugs against tuberculosis. Areas covered: This review brings to the reader the opportunity to understand the chemical and biological characteristics of all patented anti-tuberculosis drugs in North America, Europe, Japan, and Russia. The 116 patents discussed here concern new molecules in the early or advanced phase of development in the last 16 years. Expert opinion: Of all 116 patents, only one developed drug, bedaquiline, is used, and then, only in specific cases. Another three drugs are in clinical studies. However, many other compounds, for which there are in vitro and in vivo studies, seem to fulfil the requisite criteria to be a new anti-tuberculosis agent. However, why are they not in use? Why were so many studies interrupted? Why is there no more news for many of these drugs?
Collapse
Affiliation(s)
- Patricia Bento da Silva
- a Faculdade de Ciências Farmacêuticas , UNESP - Univ. Estadual Paulista, Campus Araraquara , Araraquara , São Paulo , Brazil
| | - Débora Leite Campos
- a Faculdade de Ciências Farmacêuticas , UNESP - Univ. Estadual Paulista, Campus Araraquara , Araraquara , São Paulo , Brazil
| | - Camila Maríngolo Ribeiro
- a Faculdade de Ciências Farmacêuticas , UNESP - Univ. Estadual Paulista, Campus Araraquara , Araraquara , São Paulo , Brazil
| | - Isabel Cristiane da Silva
- a Faculdade de Ciências Farmacêuticas , UNESP - Univ. Estadual Paulista, Campus Araraquara , Araraquara , São Paulo , Brazil
| | - Fernando Rogério Pavan
- a Faculdade de Ciências Farmacêuticas , UNESP - Univ. Estadual Paulista, Campus Araraquara , Araraquara , São Paulo , Brazil
| |
Collapse
|
197
|
Mikušová K, Ekins S. Learning from the past for TB drug discovery in the future. Drug Discov Today 2016; 22:534-545. [PMID: 27717850 DOI: 10.1016/j.drudis.2016.09.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 09/25/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Abstract
Tuberculosis drug discovery has shifted in recent years from a primarily target-based approach to one that uses phenotypic high-throughput screens. As examples of this, through our EU-funded FP7 collaborations, New Medicines for Tuberculosis was target-based and our more-recent More Medicines for Tuberculosis project predominantly used phenotypic screening. From these projects we have examples of success (DprE1) and failure (PimA) going from drug to target and from target to drug, respectively. It is clear that we still have much to learn about the drug targets and the complex effects of the drugs on Mycobacterium tuberculosis. We propose a more integrated approach that learns from earlier drug discovery efforts that could help to move drug discovery forward.
Collapse
Affiliation(s)
- Katarína Mikušová
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská dolina, Ilkovičova 6, 84215 Bratislava, Slovakia
| | - Sean Ekins
- Collaborative Drug Discovery, Inc., 1633 Bayshore Highway, Suite 342, Burlingame, CA 94010, USA; Collaborations in Chemistry, 5616 Hilltop Needmore Road, Fuquay Varina, NC 27526, USA.
| |
Collapse
|
198
|
Huang B, Fu J, Guo C, Wu X, Lin D, Liao X. (1)H, (15)N, (13)C resonance assignments for pyrazinoic acid binding domain of ribosomal protein S1 from Mycobacterium tuberculosis. BIOMOLECULAR NMR ASSIGNMENTS 2016; 10:321-324. [PMID: 27412769 DOI: 10.1007/s12104-016-9692-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 06/22/2016] [Indexed: 06/06/2023]
Abstract
Ribosomal protein S1 of Mycobacterium tuberculosis (MtRpsA) binds to ribosome and mRNA, and plays significant role in the regulation of translation initiation, conventional protein synthesis and transfer-messenger RNA (tmRNA) mediated trans-translation. It has been identified as the target of pyrazinoic acid (POA), a bactericidal moiety from hydrolysis of pyrazinamide, which is a mainstay of combination therapy for tuberculosis. POA prevented the interactions between the C-terminal S1 domain of MtRpsA (residues 280-368, MtRpsA(CTD)_S1) and tmRNA; so that POA can inhibit the trans-translation, which is a key component of multiple quality control pathways in bacteria. However, the details of molecular mechanism and dynamic characteristics for MtRpsA(CTD)_S1 interactions with POA, tmRNA or mRNA are still unclear. Here we present the (1)H, (15)N, (13)C resonance assignments of MtRpsA(CTD)_S1 as well as the secondary structure information based on backbone chemical shifts, which lay foundation for further solution structure determination, dynamic properties characterization and interactions investigation between MtRpsA(CTD)_S1 and tmRNA, RNA or POA.
Collapse
Affiliation(s)
- Biling Huang
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Jinglin Fu
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Chenyun Guo
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xueji Wu
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Donghai Lin
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| | - Xinli Liao
- Key Laboratory of Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
199
|
Gopal P, Yee M, Sarathy J, Low JL, Sarathy JP, Kaya F, Dartois V, Gengenbacher M, Dick T. Pyrazinamide Resistance Is Caused by Two Distinct Mechanisms: Prevention of Coenzyme A Depletion and Loss of Virulence Factor Synthesis. ACS Infect Dis 2016; 2:616-626. [PMID: 27759369 DOI: 10.1021/acsinfecdis.6b00070] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pyrazinamide (PZA) is a critical component of first- and second-line treatments of tuberculosis (TB), yet its mechanism of action largely remains an enigma. We carried out a genetic screen to isolate Mycobacterium bovis BCG mutants resistant to pyrazinoic acid (POA), the bioactive derivative of PZA, followed by whole genome sequencing of 26 POA resistant strains. Rather than finding mutations in the proposed candidate targets fatty acid synthase I and ribosomal protein S1, we found resistance conferring mutations in two pathways: missense mutations in aspartate decarboxylase panD, involved in the synthesis of the essential acyl carrier coenzyme A (CoA), and frameshift mutations in the vitro nonessential polyketide synthase genes mas and ppsA-E, involved in the synthesis of the virulence factor phthiocerol dimycocerosate (PDIM). Probing for cross resistance to two structural analogs of POA, nicotinic acid and benzoic acid, showed that the analogs share the PDIM- but not the CoA-related mechanism of action with POA. We demonstrated that POA depletes CoA in wild-type bacteria, which is prevented by mutations in panD. Sequencing 10 POA-resistant Mycobacterium tuberculosis H37Rv isolates confirmed the presence of at least 2 distinct mechanisms of resistance to the drug. The emergence of resistance through the loss of a virulence factor in vitro may explain the lack of clear molecular patterns in PZA-resistant clinical isolates, other than mutations in the prodrug-converting enzyme. The apparent interference of POA with virulence pathways may contribute to the drug's excellent in vivo efficacy compared to its modest in vitro potency.
Collapse
Affiliation(s)
- Pooja Gopal
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Michelle Yee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jickky Sarathy
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jian Liang Low
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jansy P. Sarathy
- Public Health Research Institute, Rutgers—New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Firat Kaya
- Public Health Research Institute, Rutgers—New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Véronique Dartois
- Public Health Research Institute, Rutgers—New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Martin Gengenbacher
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Thomas Dick
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| |
Collapse
|
200
|
Jung IP, Ha NR, Lee SC, Ryoo SW, Yoon MY. Development of potent chemical antituberculosis agents targeting Mycobacterium tuberculosis acetohydroxyacid synthase. Int J Antimicrob Agents 2016; 48:247-58. [DOI: 10.1016/j.ijantimicag.2016.04.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/27/2016] [Accepted: 04/30/2016] [Indexed: 10/21/2022]
|