1
|
Wang N, Meng F, Deng L, Wu L, Yang Y, Li H, Chen Y, Wei Z, Xie B, Gong L, Niu Q, Lei J, Gao J, Huang B, Wang Q, Lai X, Liu Z, Hu J. The epidemiology and gene mutation characteristics of pyrazinamide-resistant Mycobacterium tuberculosis clinical isolates in Southern China. Emerg Microbes Infect 2025; 14:2447607. [PMID: 39745172 PMCID: PMC11721771 DOI: 10.1080/22221751.2024.2447607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 12/03/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025]
Abstract
This study investigates the epidemic trend of pyrazinamide (PZA)-resistant tuberculosis in Southern China over 11 years (2012-2022) and evaluates the mutation characteristics of PZA resistance-related genes (pncA, rpsA, and panD) in clinical Mycobacterium tuberculosis (M. tuberculosis) isolates. To fulfil these goals, we analyzed the phenotypic PZA resistance characteristics of 14,927 clinical isolates for which Bactec MGIT 960 PZA drug susceptibility testing (DST) results were available, revealing that 2,054 (13.76%) isolates were resistant to PZA. After evaluating the annual variation in the PZA resistance rate among tuberculosis cases in this region, it was observed that it decreased from 37.21% to 6.45% throughout the initial 7 years (2012-2018) and then increased from 8.01% to 12.12% over the subsequent 4 years (2019-2022). Sequences of pncA were obtained from 402 clinical M. tuberculosis complex isolates. For rpsA and panD, sequences were obtained from 360 clinical M. tuberculosis complex isolates. Mutations in pncA were found in 8 out of 223 PZA-sensitive isolates (3.59%) and 105 of 179 (58.66%) PZA-resistant isolates. Conversely, non-synonymous mutations in rpsA were identified in 5 of 137 (3.65%) PZA-resistant isolates, whereas the mutation ratio of rpsA among PZA-sensitive isolates was high at 14.03% (31/221). This difference in the rpsA mutation rate was statistically significant (P = 0.001, chi-square test). No panD mutations were observed in the 137 PZA-resistant isolates, whereas two PZA-sensitive isolates harboured point mutations in panD, including one nonsense mutation (C433 T) and another C-69 T mutation. These findings indicate that rpsA and panD may not significantly contribute to the development of PZA resistance in clinical M. tuberculosis isolates.
Collapse
Affiliation(s)
- Nan Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Fanrong Meng
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Li Deng
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Ling Wu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Yu Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Hua Li
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Yuanjin Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
- Guangzhou Medical University
| | - Zeyou Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
- Guangzhou Medical University
| | - Bei Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Lan Gong
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Qun Niu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Jie Lei
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Junwen Gao
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Bo Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Qi Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Xiaomin Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
- School of public health, Sun Yat-sen University, Shen Zhen, People’s Republic of China
| | - Zhihui Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Institute of Pulmonary Diseases, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Tuberculosis, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangdong, People’s Republic of China
| |
Collapse
|
2
|
Arı O, Durmaz R, Arslanturk A. Performance of a molecular beacon-based qPCR method for detection of fluoroquinolone resistance in Mycobacterium tuberculosis: A study confirmed by sequencing. Diagn Microbiol Infect Dis 2025; 112:116865. [PMID: 40294569 DOI: 10.1016/j.diagmicrobio.2025.116865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
Multidrug-resistant tuberculosis (MDR-TB) poses a serious threat to the control and treatment of tuberculosis (TB). Rapid diagnosis of resistant strains via utilization of molecular techniques is of critical importance on the proper management of the patients. The objective of this study was to develop a quantitative polymerase chain reaction (qPCR) method for the detection of mutation-based fluoroquinolone (FQ) resistance in Mycobacterium tuberculosis isolates. Primers and a molecular-beacon probe were designed to target quinolone-resistance-determining-region (QRDR) of the gyrA gene of M. tuberculosis. Amplification conditions and concentrations of primers/probe were optimized, and the effectiveness of the optimized qPCR method was tested on 50 MDR M. tuberculosis strains. To confirm the qPCR results, all strains were also screened for mutations in the gyrA gene using Sanger sequencing. The optimized qPCR method had analytical sensitivity of 96.5 cfu/ml. The method detected FQ resistance in 7 (14 %) of the 50 MDR-strains via either no or significantly decreased fluorescence signal due to mutations associated with FQ resistance. The sequencing of these seven strains detected three resistance-associated mutations (A90V, D94A and D94G). Of the 43 FQ-susceptible strains, 10 strains with wild-type gene sequences yielded strong fluorescent signals above 450 RFU, while the remaining 33 strains harboring a non-resistance-associated mutation (S95T) showed decreased fluorescence signals <350 RFU. The sensitivity and specificity of the qPCR method for detection of the mutations related to resistance was ≥99.9 % and 98 %, respectively. Consequently, the utility of the optimized qPCR method for the identification of mutation-based FQ resistance is promising.
Collapse
Affiliation(s)
- Oguz Arı
- Central Research and Application Center, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Rıza Durmaz
- Department of Clinical Microbiology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Turkey.
| | - Ahmet Arslanturk
- National Tuberculosis Reference Laboratory, Department of Microbiology Reference Laboratories and Biological Products, General Directorate of Public Health, Ministry of Health, Ankara, Turkey
| |
Collapse
|
3
|
Chen YL, He Y, Dahl VN, Yu K, Zhang YA, Guan CP, Wang MS. Diagnostic yield of nine user-friendly bioinformatics tools for predicting Mycobacterium tuberculosis drug resistance: A systematic review and network meta-analysis. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004465. [PMID: 40258039 PMCID: PMC12011222 DOI: 10.1371/journal.pgph.0004465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/15/2025] [Indexed: 04/23/2025]
Abstract
To compare the diagnostic yield of various bioinformatics tools for predicting Mycobacterium tuberculosis drug resistance. A systematic review of PubMed, Embase, Scopus, Web of Science, CINAHL and the Cochrane Library was performed to identify studies reporting the effectiveness of bioinformatic tools for predicting resistance to anti-tuberculosis (TB) drugs. Data were collected and pooled using random-effects meta-analysis and Bayesian network meta-analysis (NMA). Summary receiver operating characteristic curves (SROCs) analysis were performed, and superiority index (SI) and area under the curve (AUC) were calculated. Thirty-three studies evaluated 9 different bioinformatics tools for predicting resistance to 14 anti-TB drugs. NMA and SROCs demonstrated that TBProfiler, TGS-TB, Mykrobe, PhyResSE, and SAM-TB all exhibited satisfactory performance. Remarkably, TBProfiler stood out with its exceptional ability to predict resistance to the majority of anti-TB drugs, including isoniazid (SI: 3.39 [95% confidence interval (CI): 0.20, 11.00]; AUC: 0.97 [0.95, 0.98]), rifampicin (SI: 6.38 [0.60, 15.00]; AUC: 0.99 [0.98, 1.00]), ethambutol (SI: 5.15 [0.60, 13.00]; AUC: 0.96 [0.94, 0.97]), streptomycin (SI: 3.67 [0.60, 11.00]; AUC: 0.97 [0.95, 0.98], amikacin (SI: 2.49 [0.14, 11.00]; AUC: 0.97 [0.96, 0.99]), kanamycin (SI: 2.26 [0.14, 9.00]; AUC: 0.98 [0.97, 0.99]), levofloxacin (SI: 1.87 [0.11, 9.00]; AUC: 0.95 [0.93, 0.97]), and prothionamide (SI: 2.73 [0.20, 7.00]; AUC: 0.87 [0.84, 0.90]). Meanwhile, Mykrobe demonstrated superior accuracy specifically for moxifloxacin (SI: 3.96 [0.11, 13.00]; AUC: 0.97 [0.95, 0.98]). Lastly, TGS-TB had the best efficacy in predicting resistance to pyrazinamide (SI: 12.53 [1.67, 17.00]; AUC: 0.97 [0.95, 0.98]), capreomycin (SI: 4.22 [0.08, 15.00]; AUC: 1.00 [0.98, 1.00]), and ethionamide (SI: 2.15 [0.33, 7.00]; AUC: 0.96 [0.94, 0.98]). TBProfiler, TGS-TB, Mykrobe, PhyResSE and SAM-TB have all demonstrated outstanding accuracy in predicting resistance to anti-TB drugs. In particular, TBProfiler stood out for its exceptional performance in predicting resistance to most anti-TB drugs, while TGS-TB excelled in predicting resistance to pyrazinamide and certain second-line drugs. The efficacy of SAM-TB requires further investigation to fully establish its reliability and effectiveness. To ensure the accuracy and reliability of genotypic drug susceptibility testing, bioinformatics tools should be refined and adapted continuously to accommodate novel and current resistance-associated mutations.
Collapse
Affiliation(s)
- Ya-Li Chen
- Department of Lab Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
- Shandong Key Laboratory of Infectious Respiratory Disease, Shandong Provincial Health Commission, Jinan, China
| | - Yu He
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | - Kan Yu
- The Marshall Centre for Infectious Diseases, Research and Training, The University of Western Australia, Perth, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Yan-An Zhang
- Shandong Key Laboratory of Infectious Respiratory Disease, Shandong Provincial Health Commission, Jinan, China
- Department of Cardiovascular Surgery, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Cui-Ping Guan
- Department of Lab Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
- Shandong Key Laboratory of Infectious Respiratory Disease, Shandong Provincial Health Commission, Jinan, China
| | - Mao-Shui Wang
- Department of Lab Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, China
- Shandong Key Laboratory of Infectious Respiratory Disease, Shandong Provincial Health Commission, Jinan, China
| |
Collapse
|
4
|
Laurent S, Phelan JE, Chindelevitch L, Walker TM, Cirillo DM, Suresh A, Rodwell TC, Miotto P, Köser CU. All parts of the WHO Mycobacterium tuberculosis mutation catalog need to be applied when evaluating its performance. Microbiol Spectr 2025:e0215724. [PMID: 40172190 DOI: 10.1128/spectrum.02157-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/14/2025] [Indexed: 04/04/2025] Open
Affiliation(s)
| | - Jody E Phelan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Leonid Chindelevitch
- MRC Center for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, United Kingdom
| | - Timothy M Walker
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Nuffield Department of Medicine, Center for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Daniela M Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Timothy C Rodwell
- FIND, Grand-Saconnex, Switzerland
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, California, USA
| | - Paolo Miotto
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudio U Köser
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Ari O, Durmaz R, Vezir S, Arslanturk A. Development of a Multiplex Real-Time PCR for Diagnosis of Tuberculosis and Multidrug Resistance. Curr Microbiol 2025; 82:219. [PMID: 40153064 DOI: 10.1007/s00284-025-04196-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/18/2025] [Indexed: 03/30/2025]
Abstract
The aim of this study was to develop a multiplex qPCR method for diagnosis of tuberculosis and multidrug resistance. The method was constructed with two multiplex qPCR mixes. Mycobacterium tuberculosis and rifampicin (RIF) resistance were assessed in the first PCR mix and isoniazid (INH) resistance in the second PCR mix. Firstly, PCR amplification conditions and concentration of primers and probes were optimized. Then, analytical sensitivity/limit of detection (LOD), analytical specificity, repeatability, accuracy, and effectiveness on clinical samples were analyzed. The LOD value was determined as 124 cfu/mL. In studies conducted on 273 clinical samples, RIF and INH resistance was detected with ≥ 99% sensitivity and specificity. No false positive or negative results were obtained in the accuracy studies. In intra-experiment, inter-experiment, and inter-lot repeatability studies, the coefficient of variation was calculated as between 0.36 and 5.3%. It was also determined that the primers/probes used in this assay did not cross-react with microorganisms other than M. tuberculosis complex. In conclusion, the method optimized in the current study has high performance characteristics indicating that it can be used as an alternative for currently available molecular methods for rapid and accurate diagnosis of pulmonary tuberculosis and multidrug resistance without needing any specific equipment. Further study with different clinical samples will be useful to provide more data regarding routine utility of this method.
Collapse
Affiliation(s)
- Oğuz Ari
- Central Research and Application Center, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Rıza Durmaz
- Department of Clinical Microbiology, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Turkey.
| | - Sedat Vezir
- Department of Clinical Microbiology, Ankara Atatürk Sanatory Education and Research Hospital, Ankara, Turkey
| | - Ahmet Arslanturk
- General Directorate of Public Health, Microbiology Reference Laboratories and Biological Products Department, National Tuberculosis Reference Laboratory, Ministry of Health, Ankara, Turkey
| |
Collapse
|
6
|
Chen Y, Zhang X, Liang J, Jiang Q, Peierdun M, Xu P, Takiff HE, Gao Q. Advantages of updated WHO mutation catalog combined with existing whole-genome sequencing-based approaches for Mycobacterium tuberculosis resistance prediction. Genome Med 2025; 17:31. [PMID: 40140944 PMCID: PMC11938600 DOI: 10.1186/s13073-025-01458-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The WHO recently released a second edition of the mutation catalog for predicting drug resistance in Mycobacterium tuberculosis (MTB). This study evaluated its effectiveness compared to existing whole-genome sequencing (WGS)-based prediction methods and proposes a novel approach for its optimization. METHODS We tested the accuracy of five tools-the WHO catalog, TB Profiler, SAM-TB, GenTB, and MD-CNN-for predicting drug susceptibility on a global dataset of 36,385 MTB isolates with high-quality phenotypic drug susceptibility testing (DST) and WGS data. By integrating the genotypic DST predictions of these five tools in an ensemble machine learning framework, we developed an improved computational model for MTB drug susceptibility prediction. We then validated the ensemble model on 860 MTB isolates with phenotypic and WGS data collected in Shenzhen, China (2013-2019) and Valencia, Spain (2014-2016). RESULTS Among the five genotypic DST tools for predicting susceptibility to ten drugs, MD-CNN exhibited the highest overall performance (AUC 92.1%; 95% CI 89.8-94.4%). The WHO catalog demonstrated the highest specificity of 97.3% (95% CI 95.8-98.4%), while TB Profiler had the best sensitivity at 79.5% (95% CI 71.8-86.2%). The ensemble machine learning model (AUC 93.4%; 95% CI 91.4-95.4%) outperformed all of the five individual tools, with a specificity of 95.4% (95% CI 93.0-97.6%) and a sensitivity of 84.1% (95% CI 78.8-88.8%), principally due to considerable improvements in second-line drug resistance predictions (AUC 91.8%; 95% CI 89.6-94.0%). CONCLUSIONS The second edition of the WHO MTB mutation catalog does not, by itself, perform better than existing tools for predicting MTB drug resistance. An integrative approach combining the WHO catalog with other genotypic DST methods significantly enhances prediction accuracy.
Collapse
Affiliation(s)
- Yiwang Chen
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Xuecong Zhang
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Jialei Liang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Qi Jiang
- School of Public Health, Public Health Research Institute of Renmin Hospital, Wuhan University, Wuhan, China
| | - Mijiti Peierdun
- Department of Epidemiology and Biostatistics, School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Peng Xu
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Howard E Takiff
- Instituto Venezolano de Investigaciones Cientificas (IVIC), Caracas, Venezuela
| | - Qian Gao
- National Clinical Research Center for Infectious Diseases, Shenzhen Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China.
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Nguyen QH, Nguyen TVA, Bañuls AL. Multi-drug resistance and compensatory mutations in Mycobacterium tuberculosis in Vietnam. Trop Med Int Health 2025. [PMID: 40078052 DOI: 10.1111/tmi.14104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
BACKGROUND Vietnam is a hotspot for the emergence and spread of multidrug-resistant Mycobacterium tuberculosis. This study aimed to perform a retrospective study on the compensatory evolution in multidrug-resistant M. tuberculosis strains and the association with drug-resistant mutations and M. tuberculosis genotypes. METHODS Hundred and seventy-three strains resistant to rifampicin (n = 126) and/or isoniazid (n = 170) (multidrug-resistant = 123) were selected according to different drug-resistant patterns and genotypes. The genes/promoter regions including rpoA, rpoB, rpoC, katG, inhA, inhA promoter, ahpC, ahpC promoter, gyrA, gyrB, and rrs were sequenced for each strain. RESULTS Frequency of rifampicin- and isoniazid-resistant mutations in multidrug-resistant strains was 99.2% and 97.0%, respectively. Mutations associated with low -high levels of drug resistance with low- or no-fitness costs compared to the wild type, including rpoB_Ser450Leu, katG_Ser315Thr, inhA-15(A-T), gyrA_Asp94Gly, and rrs_A1401GA, accounted for 46.3%, 76.4%, 16.2%, 8.9%, and 11.4%, respectively, in the multidrug-resistant strains. Beijing and Euro-American genotype strains were associated with high-level drug-resistant mutations, rpoB_Ser450Leu, katG_Ser315Thr, and gyrA_Asp94Gly, while East African-Indian genotype strains were associated with low to high-level drug-resistant mutations, rpoB_His445Asp, rpoB_His445Tyr, inhA-15(C-T) and rrs_A1401G. Multidrug-resistant strains (19.5%) harboured compensatory mutations linked to rifampicin resistance in rpoA, rpoB, or rpoC. Notably, the frequency of compensatory mutations in Beijing genotypes was significantly higher than in East African-Indian genotypes (21.1% vs. 3.3%, OR = 7.7; 95% CI = 1.0 to 61.2, p = 0.03). The proportion of multidrug-resistant strains with rpoB_Ser450Leu mutations carrying rpoA-rpoC mutations was higher than that of strains with other rpoB mutations (OR = 5.4; 95% CI = 1.4 to 21.1, p = 0.02) and was associated with Beijing strains. Only 1.2% (2/170) isoniazid-resistant strains carried aphC-52(C-T) mutation in the promoter region of the ahpC gene, which was hypothesised to be the compensatory mutation in isoniazid-resistant strains. Meanwhile, 11 isoniazid-resistant strains carried a katG mutation combined with either inhA-8(T-C) or inhA-15(A-T) mutations and were associated with East African-Indian strains. CONCLUSIONS Mutations associated with high levels of drug resistance without/with low fitness costs (rpoB_Ser450Leu and katG_Ser315Thr) along with compensatory mutations linked to rifampicin resistance were strongly associated with multidrug-resistant M. tuberculosis Beijing strains in Vietnam.
Collapse
Affiliation(s)
- Quang Huy Nguyen
- LMI DRISA, Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thi Van Anh Nguyen
- Department of Bacteriology, National Institute of Hygiene and Epidemiology (NIHE), Hanoi, Vietnam
| | - Anne-Laure Bañuls
- LMI DRISA, Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
8
|
Pal A, Mohanty D. Machine learning-based approach for identification of new resistance associated mutations from whole genome sequences of Mycobacterium tuberculosis. BIOINFORMATICS ADVANCES 2025; 5:vbaf050. [PMID: 40125545 PMCID: PMC11930343 DOI: 10.1093/bioadv/vbaf050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Motivation Currently available methods for the prediction of genotypic drug resistance in Mycobacterium tuberculosis utilize information on known markers of drug resistance. Hence, machine learning approaches are needed that can discover new resistance markers. Results Whole genome sequences with known phenotypic drug resistance profiles have been utilized to train XGBoost and ANN classifiers for 5 first-line and 8 second-line tuberculosis drugs. Benchmarking on a completely independent dataset from CRyPTIC database revealed that our method has high sensitivity (90%-95%) and specificity (94%-99%) for five first-line drugs and robust performance for six second-line drugs with a sensitivity of 77%-89% at over 95% specificity. An explainable AI method, SHapley Additive exPlanations, has successfully identified resistance mutations for each drug in a completely automated way. This approach could not only identify known resistance associated mutations in agreement with the WHO mutation catalogue, but also predicted >100 other potential resistance associated mutations for 13 antibiotics in new genes outside the known resistance loci. Identification of new resistance markers opens up the opportunity for the discovery of novel mechanisms of drug resistance. Availability and implementation Our prediction method has been implemented as TB-AMRpred webserver and command line tool, available freely at http://www.nii.ac.in/TB-AMRpred.html and https://github.com/Ankitapal1995/TB-AMRprd.
Collapse
Affiliation(s)
- Ankita Pal
- Bioinformatics Center, National Institute of Immunology, New Delhi 110067, India
| | - Debasisa Mohanty
- Bioinformatics Center, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
9
|
Guo J, Feng X, Pang J, Li W, Cai M, Cao Z, Lin F, Zhang X. Risk factors for rifampicin-susceptible and isoniazid-resistant tuberculosis in adult patients with type 2 diabetes mellitus in Nanjing. BMC Infect Dis 2025; 25:335. [PMID: 40065241 PMCID: PMC11892190 DOI: 10.1186/s12879-025-10709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE Globally, Tuberculosis(TB) with type 2 diabetes mellitus (T2DM) is becoming increasingly serious, especially the emergence of rifampicin-susceptible and isoniazid-resistant tuberculosis (Hr-TB), which increases the difficulty of treatment and the burden of disease. Therefore, this single-center retrospective cohort study analyzed risk factors of Hr-TB in adult patients with T2DM and pulmonary tuberculosis (PTB) in Nanjing to guide clinical practice and improve the long-term prognosis of patients. METHODS The clinical data of 279 adult inpatients diagnosed with culture-positive PTB and T2DM in the Second Hospital of Nanjing from January 2019 and December 2021 were collected. According to the drug susceptibility testing (DST) results, 44 patients with Hr-TB were categorized as the Hr-TB group, while the remaining 235 patients with drug-susceptible tuberculosis (DS-TB) were classified as DS-TB group. Hierarchical logistic regression was employed for multivariate analysis to identify variables associated with Hr-TB in patients with T2DM. RESULTS There were no significant differences in age, sex, body mass index (BMI), smoking, drinking, ethnicity, education level, or comorbidities between the DS-TB and Hr-TB groups. Multivariate logistic regression analysis revealed that, history of previous tuberculosis treatment (OR = 2.348, 95%CI: 1.025 ~ 5.379, P = 0.044), poor FPG control (OR = 2.402, 95%CI: 1.208 ~ 4.776, P = 0.012), and serum iron levels ≥ 14.3µmol/l (OR = 2.808, 95%CI: 1.334 ~ 5.910, P = 0.007) are independent risk factors for Hr-TB in adult patients with T2DM in Nanjing. Within the cohort, 241 patients were Newly treatment tuberculosis patients, and among them, poor FPG control (OR = 2.296, 95%CI: 1.073 ~ 4.915, P = 0.032), and serum iron levels ≥ 14.3µmol/l (OR = 2.418, 95%CI: 1.048 ~ 5.577, P = 0.038) were identified as risk factors for Hr-TB. CONCLUSION Poor fasting glycemic control and serum iron levels ≥ 14.3µmol/L are independent risk factors for the development of Hr-TB in adults with T2DM and PTB, moreover, the contribution of these as risk factors were more pronounced in the newly treatment tuberculosis patients subgroup than patients with a history of previous tuberculosis treatment. History of previous tuberculosis treatment was also found to be a risk factor for Hr-TB in adults with T2DM.
Collapse
Affiliation(s)
- Jing Guo
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Jialei Pang
- Department of Intensive Care Unit, Nanjing Chest Hospital,The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenchao Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Min Cai
- Department of Institutional Office, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China
| | - Zhiyun Cao
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China
| | - Feishen Lin
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China.
| | - Xia Zhang
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China.
| |
Collapse
|
10
|
Kulkarni SG, Laurent S, Miotto P, Walker TM, Chindelevitch L, Nathanson CM, Ismail N, Rodwell TC, Farhat MR. Multivariable regression models improve accuracy and sensitive grading of antibiotic resistance mutations in Mycobacterium tuberculosis. Nat Commun 2025; 16:2149. [PMID: 40032816 PMCID: PMC11876447 DOI: 10.1038/s41467-025-57174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
Rapid genotype-based drug susceptibility testing for the Mycobacterium tuberculosis complex (MTBC) relies on a comprehensive knowledgebase of the genetic determinants of resistance. Here we present a catalogue of resistance-associated mutations using a regression-based approach and benchmark it against the 2nd edition of the World Health Organisation (WHO) mutation catalogue. We train multivariate logistic regression models on over 52,000 MTBC isolates to associate binary resistance phenotypes for 15 antitubercular drugs with variants extracted from candidate resistance genes. Regression detects 450/457 (98%) resistance-associated variants identified using the existing method (a.k.a, SOLO method) and grades 221 (29%) more total variants than SOLO. The regression-based catalogue achieves higher sensitivity on average (+3.2 percentage points, pp) than SOLO with smaller average decreases in specificity (-1.0 pp) and positive predictive value (-1.6 pp). Sensitivity gains are highest for ethambutol, clofazimine, streptomycin, and ethionamide as regression graded considerably more resistance-associated variants than SOLO for these drugs. There is no difference between SOLO and regression with regards to meeting the target product profiles set by the WHO for genetic drug susceptibility testing, except for rifampicin, for which regression specificity is below the threshold of 98% at 97%. The regression pipeline also detects isoniazid resistance compensatory mutations in ahpC and variants linked to bedaquiline and aminoglycoside hypersusceptibility. These results inform the continued development of targeted next generation sequencing, whole genome sequencing, and other commercial molecular assays for diagnosing resistance in the MTBC.
Collapse
Affiliation(s)
- Sanjana G Kulkarni
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sacha Laurent
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Paolo Miotto
- IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Timothy M Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Leonid Chindelevitch
- Medical Research Council (MRC) Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | | | - Nazir Ismail
- Global Tuberculosis Programme, World Health Organization (WHO), Geneva, Switzerland
- Department of Clinical Microbiology and Infectious Diseases, University of the Witwatersrand, Johannesburg, South Africa
| | - Timothy C Rodwell
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, CA, USA
| | - Maha R Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Division of Pulmonary & Critical Care, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
11
|
Marais BJ, Zhang X, Sintchenko V. The promise and the reality of targeted next-generation sequencing for drug-resistant tuberculosis detection. THE LANCET. INFECTIOUS DISEASES 2025; 25:251-253. [PMID: 39486427 DOI: 10.1016/s1473-3099(24)00602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 11/04/2024]
Affiliation(s)
- Ben J Marais
- The Sydney Infectious Diseases Institute (Sydney ID), University of Sydney, Sydney, NSW 2145, Australia.
| | - Xiaomei Zhang
- The Sydney Infectious Diseases Institute (Sydney ID), University of Sydney, Sydney, NSW 2145, Australia
| | - Vitali Sintchenko
- The Sydney Infectious Diseases Institute (Sydney ID), University of Sydney, Sydney, NSW 2145, Australia; NSW Mycobacterium Reference Laboratory, Centre for Infectious Diseases and Microbiology-Public Health (CIDM-PH), Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Sydney, NSW, Australia
| |
Collapse
|
12
|
Munro JE, Coussens AK, Bahlo M. TBtypeR: Sensitive detection and sublineage classification of Mycobacterium tuberculosis complex mixed-strain infections. Commun Biol 2025; 8:260. [PMID: 39972208 PMCID: PMC11840096 DOI: 10.1038/s42003-025-07705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
Tuberculosis (TB) is typically attributed to a single infecting strain of the Mycobacterium tuberculosis Complex (MTBC), however mixed-strain infections are more common than reported due to the limitations of conventional diagnostics. While whole genome sequencing (WGS) methods have improved the detection of mixed-strain infections, existing tools struggle to reliably identify mixed-strain infections with frequencies below 10%. TBtypeR, a new tool, addresses this challenge by comparing WGS data to a phylogenetic SNP panel of over 10,000 sites and 164 MTBC phylotypes and using a model based on the binomial distribution to classify sublineage mixtures at frequencies as low as 1%. Extensive benchmarking shows TBtypeR outperforms current methods. Application to a published dataset of 5000 WGS samples identified 305 mixed-strain infections, six-fold higher than previously reported. The TBtypeR R package and a Nextflow pipeline are available at github.com/bahlolab/TBtypeR, providing a powerful tool for studying TB epidemiology and mixed-strain infections.
Collapse
Affiliation(s)
- Jacob E Munro
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, 3052, Australia.
| | - Anna K Coussens
- Department of Medical Biology, University of Melbourne, Parkville, 3052, Australia
- Infection and Global Health Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, Republic of South Africa
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, 3052, Australia
| |
Collapse
|
13
|
Li K, Li M, Liu W, Wu Y, Li F, Xie J, Zhou S, Wang S, Guo Y, Pan J, Wang X. Electroencephalographic differences between waking and sleeping periods in patients with prolonged disorders of consciousness at different levels of consciousness. Front Hum Neurosci 2025; 19:1521355. [PMID: 40034215 PMCID: PMC11872887 DOI: 10.3389/fnhum.2025.1521355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Objective This study aimed to explore differences in sleep electroencephalogram (EEG) patterns in individuals with prolonged disorders of consciousness, utilizing polysomnography (PSG) to assist in distinguishing between the vegetative state (VS)/unresponsive wakefulness syndrome (UWS) and the minimally conscious state (MCS), thereby reducing misdiagnosis rates and enhancing the quality of medical treatment. Methods A total of 40 patients with prolonged disorders of consciousness (pDOC; 27 patients in the VS/UWS and 13 in the MCS) underwent polysomnography. We analyzed differential EEG indices between VS/UWS and MCS groups and performed correlation analyses between these indices and the Coma Recovery Scale-Revised (CRS-R) scores. The diagnostic accuracy of the differential indices was evaluated using receiver operating characteristic (ROC) curves. Results 1. The fractal dimension (Higuchi's fractal dimension (HFD)) of patients in the MCS tended to be higher than that of patients in the VS/UWS across all phases, with a significant difference only in the waking phase (p < 0.05). The HFD in the waking phase was positively correlated with the CRS-R score and exhibited the highest diagnostic accuracy at 88.3%. The Teager-Kaiser energy operator (TKEO) also showed higher levels in patients in the MCS compared to those in the VS/UWS, significantly so in the NREM2 phase (p < 0.05), with a positive correlation with the CRS-R score and diagnostic accuracy of 75.2%. The δ-band power spectral density [PSD(δ)] in the patients in the MCS was lower than that in those in the VS/UWS, significantly so in the waking phase (p < 0.05), and it was negatively correlated with the CRS-R score, with diagnostic accuracy of 71.5%. Conclusion Polysomnography for the VS/UWS and MCS revealed significant differences, aiding in distinguishing between the two patient categories and reducing misdiagnosis rates. Notably, the HFD and PSD(δ) showed significantly better performance during wakefulness compared to sleep, while the TKEO was more prominent in the NREM2 stage. Notably, the HFD exhibited a robust correlation with the CRS-R scores, the highest diagnostic accuracy, and immense promise in the clinical diagnosis of prolonged disorders of consciousness.
Collapse
Affiliation(s)
- Keke Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Man Li
- School of Automation Science and Engineering, South China University of Technology, Guangzhou, China
| | - Wanqing Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanzhi Wu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Li
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingwei Xie
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Shaolong Zhou
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Sen Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongkun Guo
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| | - Jiahui Pan
- School of Software, South China Normal University, Nanhai Software Technology Park, Foshan, Guangdong Province, China
| | - Xinjun Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Neurosurgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Research Center for Prevention and Treatment of Brain Injuries, Zhengzhou, China
- Henan Key Laboratory of Brain Science and Brain Computer Interface Technology, Zhengzhou, China
| |
Collapse
|
14
|
Kalinich CC, Gonzalez FL, Osmaston A, Breban MI, Distefano I, Leon C, Sheen P, Zimic M, Coronel J, Tan G, Crudu V, Ciobanu N, Codreanu A, Solano W, Ráez J, Allicock OM, Chaguza C, Wyllie AL, Brandt M, Weinberger DM, Sobkowiak B, Cohen T, Grandjean L, Grubaugh ND, Redmond SN. Tiled Amplicon Sequencing Enables Culture-free Whole-Genome Sequencing of Pathogenic Bacteria From Clinical Specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629550. [PMID: 39763738 PMCID: PMC11702625 DOI: 10.1101/2024.12.19.629550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Pathogen sequencing is an important tool for disease surveillance and demonstrated its high value during the COVID-19 pandemic. Viral sequencing during the pandemic allowed us to track disease spread, quickly identify new variants, and guide the development of vaccines. Tiled amplicon sequencing, in which a panel of primers is used for multiplex amplification of fragments across an entire genome, was the cornerstone of SARS-CoV-2 sequencing. The speed, reliability, and cost-effectiveness of this method led to its implementation in academic and public health laboratories across the world and adaptation to a broad range of viral pathogens. However, similar methods are not available for larger bacterial genomes, for which whole-genome sequencing typically requires in vitro culture. This increases costs, error rates and turnaround times. The need to culture poses particular problems for medically important bacteria such as Mycobacterium tuberculosis, which are slow to grow and challenging to culture. As a proof of concept, we developed two novel whole-genome amplicon panels for M. tuberculosis and Streptococcus pneumoniae. Applying our amplicon panels to clinical samples, we show the ability to classify pathogen subgroups and to reliably identify markers of drug resistance without culturing. Development of this work in clinical settings has the potential to dramatically reduce the time of diagnosis of drug resistance for multiple drugs in parallel, enabling earlier intervention for high priority pathogens.
Collapse
Affiliation(s)
- Chaney C Kalinich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Freddy L Gonzalez
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
| | - Alice Osmaston
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mallery I Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Isabel Distefano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Candy Leon
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Mirko Zimic
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Grace Tan
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
| | | | | | | | | | - Jimena Ráez
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Orchid M Allicock
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
| | - Chrispin Chaguza
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anne L Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Matthew Brandt
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Benjamin Sobkowiak
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Louis Grandjean
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Seth N Redmond
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Wang S, Xiao X, Dong S, Cao J, Wang S, Xiong H, Li X, Shao G, Hu Y, Shen X. Analysis of genetic characteristics associated with reduced bedaquiline susceptibility in multidrug-resistant Mycobacterium tuberculosis. Tuberculosis (Edinb) 2024; 149:102572. [PMID: 39504872 DOI: 10.1016/j.tube.2024.102572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/12/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024]
Abstract
Bedaquiline (BDQ) has shown efficacy in shortening treatment duration and enhancing treatment success rates for multidrug-resistant tuberculosis (MDR-TB), thereby prompting widespread adoption. However, resistance to BDQ has emerged. This study aimed to identify genetic characteristics associated with decreased susceptibility to BDQ, using a public database to aid in the detection of resistant strains. Seventy-one BDQ-resistant and 929 BDQ-susceptible isolates from the open-source CRyPTIC database were selected for analysis. Variant calling was conducted via the clockwork pipeline. Univariate logistic regression was performed for each gene mutation, followed by LASSO regression for further variant selection. Ultimately, a multiple linear regression model was developed using log2-transformed Minimum Inhibitory Concentration values as the dependent variable, with variant selection refined through stepwise regression based on the Akaike Information Criterion. Ten gene mutations were significantly associated with reduced BDQ susceptibility, including two key gene mutations: Rv0678_141_ins_1 and Rv1979c_D249E, with effect estimates of 1.76 (95 % CI: 0.67-2.84) and 1.69 (95 % CI: 0.22-3.17), respectively. Other implicated genes included Rv2699c_-84_del_1, hsaB_I179T, mmpL9_T241A, pncA_C14R, Rv0373c_G621S, Rv0893c_L27F, Rv1770_A4D, and Rv3428c_S327C. This study identified ten gene mutations linked to decreased susceptibility to BDQ, providing a reference for developing a comprehensive catalog of BDQ-resistant genes.
Collapse
Affiliation(s)
- Shanshan Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Xiao Xiao
- Division of TB and HIV/AIDS Prevention, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, 200336, China.
| | - Shulan Dong
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Jiayi Cao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Sainan Wang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Haiyan Xiong
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Xuliang Li
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Ge Shao
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Yi Hu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China; Key Laboratory of Public Health Safety (Fudan University), Ministry of Education, China.
| | - Xin Shen
- Division of TB and HIV/AIDS Prevention, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, 200336, China.
| |
Collapse
|
16
|
Li Y, Shao Y, Li Y, Kong X, Tao N, Hou Y, Wang T, Li Y, Liu Y, Li H. Association between toxin-antitoxin system mutations and global transmission of MDR-TB. BMC Infect Dis 2024; 24:1250. [PMID: 39501228 PMCID: PMC11539496 DOI: 10.1186/s12879-024-10142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The emergence of Multidrug-Resistant Tuberculosis (MDR-TB) poses a significant threat to global tuberculosis control efforts. This study aimed to examine the influence of mutations in Toxin-Antitoxin system genes on the global transmission of MDR-TB caused by Mycobacterium tuberculosis (M. tuberculosis). METHODS Whole-genome sequencing was conducted on 13,518 M. tuberculosis isolates. Genes of the Toxin-Antitoxin system were obtained from the National Center for Biotechnology Information (NCBI) Gene database. Techniques such as Random Forest, Gradient Boosting Decision Tree, and Generalized Linear Mixed Models were employed to identify mutation sites in Toxin-Antitoxin system-related genes that facilitated the transmission of MDR-TB. RESULTS 4,066 (30.08%) were identified as MDR-TB strains of all analyzed isolates. We found significant associations between specific gene mutations and MDR-TB transmission clusters including mutations in Rv0298 (G213A), Rv1959c (parE1, C88T), Rv1960c (parD1, C134T), Rv1991A (maze, G156A), Rv2547 (vapB, C54G), Rv2862A (vapB23, T2C), and Rv3385c (vapB46, G70A). Additionally, several gene mutations associated with MDR-TB transmission clades such as Rv1956 (higA, G445T), Rv1960c (parD1, C134T), and Rv1962A (vapB35, G99A) were noted. Certain gene mutations including vapB35 (G99A), higA (G445T), and parD1 (C134T) correlated with cross-regional transmission clades. CONCLUSION This study highlights the significant association between specific gene mutations in the Toxin-Antitoxin system and the global transmission of MDR-TB, providing valuable insights for developing targeted interventions to control MDR-TB.
Collapse
Affiliation(s)
- Yameng Li
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- College of the First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yang Shao
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yifan Li
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Shandong First Medical University (Affiliated Hospital of Shandong Academy of Medical Sciences), Jinan, 250031, Shandong, China
| | - Xianglong Kong
- Artificial Intelligence Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250011, Shandong, China
| | - Ningning Tao
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yawei Hou
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Tingting Wang
- College of the First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yingying Li
- College of the First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yao Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Huaichen Li
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- College of the First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
17
|
Pei S, Song Z, Yang W, He W, Ou X, Zhao B, He P, Zhou Y, Xia H, Wang S, Jia Z, Walker TM, Zhao Y. The catalogue of Mycobacterium tuberculosis mutations associated with drug resistance to 12 drugs in China from a nationwide survey: a genomic analysis. THE LANCET. MICROBE 2024; 5:100899. [PMID: 39353459 PMCID: PMC11543636 DOI: 10.1016/s2666-5247(24)00131-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND WHO issued the first edition catalogue of Mycobacterium tuberculosis complex (MTBC) mutations associated with drug resistance in 2021. However, country-specific issues might lead to arising complex and additional drug-resistant mutations. We aimed to fully reflect the characteristics of drug resistance mutations in China. METHODS We analysed MTBC isolates from the nationwide drug-resistant tuberculosis surveillance with 70 counties in 31 provinces, municipalities, and autonomous regions in China. Three types of MYCOTB plates were used to perform drug susceptibility testing for 12 antibiotics (rifampicin, isoniazid, ethambutol, levofloxacin, moxifloxacin, amikacin, kanamycin, ethionamide, clofazimine, linezolid, delamanid, and bedaquiline). Mutations were divided into five groups according to their odds ratios, positive predictive values, false discovery rate-corrected p values, and 95% CIs: (1) associated with resistance; (2) associated with resistance-interim; (3) uncertain significance; (4) not associated with resistance-interim; and (5) not associated with resistance. The Wilcoxon rank-sum and Kruskal-Wallis tests were used to quantify the association between mutations and minimum inhibitory concentrations (MICs). Our dataset was compared with the first edition of the WHO catalogue. FINDINGS We analysed 10 146 MTBC isolates, of which 9071 (89·4%) isolates were included in the final analysis. 744 (8·2%) isolates were resistant to rifampicin and 1339 (14·8%) to isoniazid. 208 (1·9%) of 11 065 mutations were classified as associated with resistance or associated with resistance-interim. 33 (97·1%) of 34 mutations in group 1 and 92 (52·9%) of 174 in group 2 also appeared in groups 1 or 2 of the WHO catalogue. Of 81 indel mutations in group 2, 15 (18·5%) were in the WHO catalogue. The newly discovered mutation gyrA_Ala288Asp was associated with levofloxacin resistance. MIC values for rifampicin, isoniazid, moxifloxacin, and levofloxacin corresponding to resistance mutations in group 1 were significantly different (p<0·0001), and 12 high-level resistance mutations were detected. 61 mutations in group 3 occurred as solo in at least five phenotypically susceptible isolates, but with MIC values moderately higher than other susceptible isolates. Among 945 phenotypically resistant but genotypically susceptible isolates, 433 (45·8%) were mutated for at least one efflux pump gene. INTERPRETATION Our analysis reflects the complexity of drug resistance mutations in China and suggests that indel mutations, efflux pump genes, protein structure, and MICs should be fully considered in the WHO catalogue, especially in countries with a high tuberculosis burden. FUNDING National Key Research and Development Program of China and the Science and Technology Major Project of Tibetan Autonomous Region of China.
Collapse
Affiliation(s)
- Shaojun Pei
- Department of Global Health, School of Public Health, Peking University, Beijing, China; National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zexuan Song
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wei Yang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Wencong He
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xichao Ou
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bing Zhao
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ping He
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Zhou
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Xia
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shengfen Wang
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhongwei Jia
- Department of Global Health, School of Public Health, Peking University, Beijing, China.
| | - Timothy M Walker
- Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam; Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
| | - Yanlin Zhao
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Chinese Center for Disease Control and Prevention, Beijing, China; National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China.
| |
Collapse
|
18
|
Lipworth S, Crook D, Walker AS, Peto T, Stoesser N. Exploring uncatalogued genetic variation in antimicrobial resistance gene families in Escherichia coli: an observational analysis. THE LANCET. MICROBE 2024; 5:100913. [PMID: 39378891 PMCID: PMC7617469 DOI: 10.1016/s2666-5247(24)00152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Antimicrobial resistance (AMR) in Escherichia coli is a global problem associated with substantial morbidity and mortality. AMR-associated genes are typically annotated based on similarity to variants in a curated reference database, with the implicit assumption that uncatalogued genetic variation within these is phenotypically unimportant. In this study, we evaluated the performance of the AMRFinder tool and, subsequently, the potential for discovering new AMR-associated gene families and characterising variation within existing ones to improve genotype-to-susceptibility phenotype predictions in E coli. METHODS In this cross-sectional study of international genome sequence data, we assembled a global dataset of 9001 E coli sequences from five publicly available data collections predominantly deriving from human bloodstream infections from: Norway, Oxfordshire (UK), Thailand, the UK, and Sweden. 8555 of these sequences had linked antibiotic susceptibility data. Raw reads were assembled using Shovill and AMR genes (relevant to amoxicillin-clavulanic acid, ampicillin, ceftriaxone, ciprofloxacin, gentamicin, piperacillin-tazobactam, and trimethoprim) extracted using the National Center for Biotechnology Information AMRFinder tool (using both default and strict [100%] coverage and identity filters). We assessed the predictive value of the presence of these genes for predicting resistance or susceptibility against US Food and Drug Administration thresholds for major and very major errors. Mash was used to calculate the similarity between extracted genes using Jaccard distances. We empirically reclustered extracted gene sequences into AMR-associated gene families (≥70% match) and antibiotic-resistance genes (ARGs; 100% match) and categorised these according to their frequency in the dataset. Accumulation curves were simulated and correlations between gene frequency in the Oxfordshire and other datasets calculated using the Spearman coefficient. Firth regression was used to model the association between the presence of blaTEM-1 variants and amoxicillin-clavulanic acid or piperacillin-tazobactam resistance, adjusted for the presence of other relevant ARGs. FINDINGS The performance of the AMRFinder database for genotype-to-phenotype predictions using strict 100% identity and coverage thresholds did not meet US Food and Drug Administration thresholds for any of the seven antibiotics evaluated. Relaxing filters to default settings improved sensitivity with a specificity cost. For all antibiotics, most explainable resistance was associated with the presence of a small number of genes. There was a proportion of resistance that could not be explained by known ARGs; this ranged from 75·1% for amoxicillin-clavulanic acid to 3·4% for ciprofloxacin. Only 18 199 (51·5%) of the 35 343 ARGs detected had a 100% identity and coverage match in the AMRFinder database. After empirically reclassifying genes at 100% nucleotide sequence identity, we identified 1042 unique ARGs, of which 126 (12·1%) were present ten times or more, 313 (30·0%) were present between two and nine times, and 603 (57·9%) were present only once. Simulated accumulation curves revealed that discovery of new (100% match) ARGs present more than once in the dataset plateaued relatively quickly, whereas new singleton ARGs were discovered even after many thousands of isolates had been included. We identified a strong correlation (Spearman coefficient 0·76 [95% CI 0·73-0·80], p<0·0001) between the number of times an ARG was observed in Oxfordshire and the number of times it was seen internationally, with ARGs that were observed six times in Oxfordshire always being found elsewhere. Finally, using the example of blaTEM-1, we showed that uncatalogued variation, including synonymous variation, is associated with potentially important phenotypic differences; for example, two common, uncatalogued blaTEM-1 alleles with only synonymous mutations compared with the known reference were associated with reduced resistance to amoxicillin-clavulanic acid (adjusted odds ratio 0·58 [95% CI 0·35-0·95], p=0·031) and piperacillin-tazobactam (0·50 [95% CI 0·29-0·82], p=0·005). INTERPRETATION We highlight substantial uncatalogued genetic variation with respect to known ARGs, although a relatively small proportion of these alleles are repeatedly observed in a large international dataset suggesting strong selection pressures. The current approach of using fuzzy matching for ARG detection, ignoring the unknown effects of uncatalogued variation, is unlikely to be acceptable for future clinical deployment. The association of synonymous mutations with potentially important phenotypic differences suggests that relying solely on amino acid-based gene detection to predict resistance is unlikely to be sufficient. Finally, the inability to explain all resistance using existing knowledge highlights the importance of new target gene discovery. FUNDING National Institute for Health and Care Research, Wellcome, and UK Medical Research Council.
Collapse
Affiliation(s)
- Samuel Lipworth
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Derrick Crook
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| | - A Sarah Walker
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| | - Tim Peto
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Nicole Stoesser
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at University of Oxford in partnership with UKHSA, Oxford, UK
| |
Collapse
|
19
|
Ou X, Song Z, Xing R, Zhao B, Pei S, Teng C, Zhang L, Sun Q, Liu F, Xia H, Zhou Y, Zheng Y, Song Y, Zhang Z, Wang S, Anthony R, Zhao Y. Development and preliminary assessment of the iFIND TBR: all-in- one molecular diagnostic assay for rapid detection of Mycobacterium tuberculosis and rifampicin resistance. Front Cell Infect Microbiol 2024; 14:1439099. [PMID: 39534701 PMCID: PMC11554655 DOI: 10.3389/fcimb.2024.1439099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Early and accurate diagnosis of tuberculosis (TB) is crucial for initiating timely treatment and preventing new infections. In this study, we introduced the iFIND TBR assay, an automated all-in-one tuberculosis detection approach that simultaneously detect Mycobacterium tuberculosis (MTB) and rifampicin (RIF) resistance. Methods The limits of detection (LOD), sensitivity, specificity, and RIF-R rpoB mutation detection of the iFIND TBR were tested on Mycobacterium tuberculosis DNA or sputum samples spiked with known numbers of M.tuberculosis H37Rv. Frozen clinical samples from patients suspected of having TB were also tested. Results The LOD of the iFIND TBR for MTB detection were 13.34 CFU/ml (95% CI, 11.71-16.47), and for RIF resistance was 109.79CFU/mL (95% CI, 95-138.19). The iFIND TBR assay accurately distinguish MTB strains from non-tuberculous mycobacteria (NTM) without any cross reactivity. Testing on 157 clinical sputum samples, compared with the bacteriologically TB standard, the overall sensitivity and specificity of the iFIND TBR was 100% (95%CI, 94.64, 100) and 85.29% (95% CI, 74.61, 92.72), respectively. When assessing RIF susceptibility, the iFIND TBR achieved a sensitivity of 98.15% (95% CI, 90.11-99.95) and a specificity of 85.71% (95% CI, 67.33-95.97), compared with phenotypic drug susceptibility testing. Discordant RIF susceptibility results were more frequently observed in samples exhibiting heteroresistance. Discussion These findings demonstrate that iFIND TBR assay performs well in detecting TB and RIF resistance, and shows promise as a point-of-care tool in resource-limited areas.
Collapse
Affiliation(s)
- Xichao Ou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zexuan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Clinical Laboratory, Children’s Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Ruida Xing
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Bing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shaojun Pei
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Public Health, Peking University, Beijing, China
| | - Chong Teng
- Department of Tuberculosis, Beijing Dongcheng District Center for Disease Control and Prevention, Beijing, China
| | - Lincai Zhang
- Institute for Tuberculosis Control and Prevention, Gansu Provincial Center for Disease Control and Prevention, Lanzhou, China
| | - Qian Sun
- Department of Clinical Laboratory, Tuberculosis Dispensary of Changping District, Beijing, China
| | - Fang Liu
- Institute for Tuberculosis Control and Prevention, Gansu Provincial Center for Disease Control and Prevention, Lanzhou, China
| | - Hui Xia
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Zheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuanyuan Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhiguo Zhang
- Department of Clinical Laboratory, Tuberculosis Dispensary of Changping District, Beijing, China
| | - Shengfen Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Richard Anthony
- National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Yanlin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
20
|
Nandu N, Miller M, Tong Y, Lu ZX. A novel dual probe-based method for mutation detection using isothermal amplification. PLoS One 2024; 19:e0309541. [PMID: 39436873 PMCID: PMC11495626 DOI: 10.1371/journal.pone.0309541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/13/2024] [Indexed: 10/25/2024] Open
Abstract
Cost efficient and rapid detection tools to detect mutations especially those linked to drug-resistance are important to address concerns of the rising multi-drug resistance infections. Here we integrated dual probes, namely a calibrator probe and an indicator probe, into isothermal amplification detection system. These two probes are designed to bind distinct regions on the same amplicon to determine the presence or absence of mutation. The calibrator probe signal is used as an internal signal calibrator for indicator probe which detects the presence or absence of the mutation. As an illustrative example, we evaluated the applicability of this dual probe method for detecting mutations associated with rifampicin (RIF) drug resistance at codons 516, 526 and 531 of the rpoB gene in Mycobacterium tuberculosis. In this assessment, we examined 127 artificial samples comprising wild types and mutants with single or multiple mutations. Our results demonstrated 100% accuracy for both wild types and mutants for mutations at codons 526 and 531. As regards to mutations at codon 516, the wild type was identified with 100% accuracy, while the mutants were identified with 95% accuracy. Moreover, when we extended our evaluation to include clinical MTB strains and the Zeptometrix MTB Verification panel, our method achieved 100% accuracy (5 out of 5) in identifying wild-type strains. Additionally, we successfully detected a RIF-resistant strain with mutations at codon 531 of the rpoB gene in Zeptometrix verification panel. Our isothermal mutation detection system, relying on dual probes exhibits a versatile approach. With the capability to identify mutations without prior knowledge of their specific mutation direction, our dual-probe method shows significant promise for applications in drug resistance nucleic acid testing, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Nidhi Nandu
- Revvity, Inc., Waltham, MA, United States of America
| | | | - Yanhong Tong
- Revvity, Inc., Waltham, MA, United States of America
| | - Zhi-xiang Lu
- Revvity, Inc., Waltham, MA, United States of America
| |
Collapse
|
21
|
Cabibbe AM, Moghaddasi K, Batignani V, Morgan GSK, Di Marco F, Cirillo DM. Nanopore-based targeted sequencing test for direct tuberculosis identification, genotyping, and detection of drug resistance mutations: a side-by-side comparison of targeted next-generation sequencing technologies. J Clin Microbiol 2024; 62:e0081524. [PMID: 39240079 PMCID: PMC11481573 DOI: 10.1128/jcm.00815-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
We investigated the performance of the targeted next-generation sequencing (tNGS)-based Oxford Nanopore Diagnostics AmPORE TB assay, recently approved by the World Health Organization (WHO) as tuberculosis (TB) diagnostic test for the detection of drug resistance on respiratory specimens. A total of 104 DNA samples from Xpert MTB/RIF-positive TB sputum specimens were tested using the AmPORE TB kit, with the GenoScreen Deeplex Myc-TB as a comparative tNGS assay. For AmPORE TB, DNA samples were divided into five sequencing runs on the MinION device. Data analysis was performed using proprietary software. The WHO catalog of mutations was used for drug resistance interpretation. The assay achieved a high validity rate of 98% (102/104 DNA samples), homogeneous mean reads coverage across TB-positive specimens, and 100% positive and negative agreements for detecting mutations associated with resistance to rifampicin, pyrazinamide, fluoroquinolones, ethambutol, and capreomycin compared with Deeplex Myc-TB. The main discrepancies for the remaining drugs were attributable to the different assay panel designs. The AmPORE TB turnaround time was approximately 5-6 hours from extracted DNA to tNGS reporting for batches of 22 DNA samples. The AmPORE TB assay drastically reduced the time to tNGS reporting from days to hours and showed good performance for drug-resistant TB profiling compared with Deeplex Myc-TB. IMPORTANCE Targeted next-generation sequencing (tNGS) of Mycobacterium tuberculosis provides comprehensive resistance predictions matched to new multidrug-resistant/rifampicin-resistant tuberculosis regimens and received World Health Organization approval for clinical use in respiratory samples in 2024. The advanced version of the Oxford Nanopore Diagnostics AmPORE TB tNGS kit was evaluated in this study for the first time and demonstrated good performance, flexibility, and faster turnaround time compared with the existing solutions.
Collapse
Affiliation(s)
| | - Kiarash Moghaddasi
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Virginia Batignani
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Federico Di Marco
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Maria Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Hall MB, Wick RR, Judd LM, Nguyen AN, Steinig EJ, Xie O, Davies M, Seemann T, Stinear TP, Coin L. Benchmarking reveals superiority of deep learning variant callers on bacterial nanopore sequence data. eLife 2024; 13:RP98300. [PMID: 39388235 PMCID: PMC11466455 DOI: 10.7554/elife.98300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Variant calling is fundamental in bacterial genomics, underpinning the identification of disease transmission clusters, the construction of phylogenetic trees, and antimicrobial resistance detection. This study presents a comprehensive benchmarking of variant calling accuracy in bacterial genomes using Oxford Nanopore Technologies (ONT) sequencing data. We evaluated three ONT basecalling models and both simplex (single-strand) and duplex (dual-strand) read types across 14 diverse bacterial species. Our findings reveal that deep learning-based variant callers, particularly Clair3 and DeepVariant, significantly outperform traditional methods and even exceed the accuracy of Illumina sequencing, especially when applied to ONT's super-high accuracy model. ONT's superior performance is attributed to its ability to overcome Illumina's errors, which often arise from difficulties in aligning reads in repetitive and variant-dense genomic regions. Moreover, the use of high-performing variant callers with ONT's super-high accuracy data mitigates ONT's traditional errors in homopolymers. We also investigated the impact of read depth on variant calling, demonstrating that 10× depth of ONT super-accuracy data can achieve precision and recall comparable to, or better than, full-depth Illumina sequencing. These results underscore the potential of ONT sequencing, combined with advanced variant calling algorithms, to replace traditional short-read sequencing methods in bacterial genomics, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Michael B Hall
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Ryan R Wick
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Centre for Pathogen Genomics, The University of MelbourneMelbourneAustralia
| | - Louise M Judd
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Centre for Pathogen Genomics, The University of MelbourneMelbourneAustralia
| | - An N Nguyen
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Eike J Steinig
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Ouli Xie
- Department of Infectious Diseases, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Monash Infectious Diseases, Monash HealthMelbourneAustralia
| | - Mark Davies
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Torsten Seemann
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Centre for Pathogen Genomics, The University of MelbourneMelbourneAustralia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Centre for Pathogen Genomics, The University of MelbourneMelbourneAustralia
| | - Lachlan Coin
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| |
Collapse
|
23
|
Farhat M, Cox H, Ghanem M, Denkinger CM, Rodrigues C, Abd El Aziz MS, Enkh-Amgalan H, Vambe D, Ugarte-Gil C, Furin J, Pai M. Drug-resistant tuberculosis: a persistent global health concern. Nat Rev Microbiol 2024; 22:617-635. [PMID: 38519618 DOI: 10.1038/s41579-024-01025-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 03/25/2024]
Abstract
Drug-resistant tuberculosis (TB) is estimated to cause 13% of all antimicrobial resistance-attributable deaths worldwide and is driven by both ongoing resistance acquisition and person-to-person transmission. Poor outcomes are exacerbated by late diagnosis and inadequate access to effective treatment. Advances in rapid molecular testing have recently improved the diagnosis of TB and drug resistance. Next-generation sequencing of Mycobacterium tuberculosis has increased our understanding of genetic resistance mechanisms and can now detect mutations associated with resistance phenotypes. All-oral, shorter drug regimens that can achieve high cure rates of drug-resistant TB within 6-9 months are now available and recommended but have yet to be scaled to global clinical use. Promising regimens for the prevention of drug-resistant TB among high-risk contacts are supported by early clinical trial data but final results are pending. A person-centred approach is crucial in managing drug-resistant TB to reduce the risk of poor treatment outcomes, side effects, stigma and mental health burden associated with the diagnosis. In this Review, we describe current surveillance of drug-resistant TB and the causes, risk factors and determinants of drug resistance as well as the stigma and mental health considerations associated with it. We discuss recent advances in diagnostics and drug-susceptibility testing and outline the progress in developing better treatment and preventive therapies.
Collapse
Affiliation(s)
- Maha Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Helen Cox
- Institute of Infectious Disease and Molecular Medicine, Wellcome Centre for Infectious Disease Research and Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa
| | - Marwan Ghanem
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Claudia M Denkinger
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Heidelberg University Hospital, Heidelberg, Germany
| | | | - Mirna S Abd El Aziz
- Division of Infectious Disease and Tropical Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Debrah Vambe
- National TB Control Programme, Manzini, Eswatini
| | - Cesar Ugarte-Gil
- School of Public and Population Health, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer Furin
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA
| | - Madhukar Pai
- McGill International TB Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
24
|
Dong Y, Ou X, Zhao B, Wang Y, Liu Y, Liu Z, Wang H, Ge X, Nan Y, Zhao Y, Zhou X. Phylogenetically Informative Mutations in Drug Resistance Genes of Human-Infecting Mycobacterium bovis. Transbound Emerg Dis 2024; 2024:5578214. [PMID: 40303120 PMCID: PMC12017247 DOI: 10.1155/2024/5578214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/03/2024] [Accepted: 08/08/2024] [Indexed: 05/02/2025]
Abstract
The diagnosis of drug-resistant tuberculosis (TB) by molecular testing of Mycobacterium tuberculosis drug resistance genes is becoming increasingly common clinically. However, M. bovis, as an uncommon pathogen of human TB, may interfere with the test results. A comprehensive understanding of phylogenetically informative mutations in the drug resistance genes of M. bovis is required to distinguish true resistance-conferring mutations. We analyzed 53 drug resistance genes in 165 M. bovis isolated from humans using whole-genome sequencing data and found that 98.2% (162/165) of isolates have pyrazinamide intrinsic genotypic resistance, owing to the H57D mutation in the pncA gene. 12.1% (20/165) of M. bovis isolates were resistant to drugs other than pyrazinamide. Furthermore, we discovered 18 phylogenetically informative mutations that differed between M. bovis and the major lineages 1-4 of M. tuberculosis. Additionally, we reported false-positive ethambutol resistance caused by M. bovis infection due to the phylogenetically informative mutation embB E378A. This study is crucial for gaining insights into the genetic characterization and drug resistance of M. bovis prevalent in humans, as well as contributing to the development of more accurate molecular diagnostic methods and detection tools for drug resistance.
Collapse
Affiliation(s)
- Yuhui Dong
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Xichao Ou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious DiseasesNational Center for Tuberculosis Control and PreventionChinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Bing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious DiseasesNational Center for Tuberculosis Control and PreventionChinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yuanzhi Wang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Yiduo Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Ziyi Liu
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Haoran Wang
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Xin Ge
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Yue Nan
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| | - Yanlin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious DiseasesNational Center for Tuberculosis Control and PreventionChinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiangmei Zhou
- National Key Laboratory of Veterinary Public Health and SafetyCollege of Veterinary MedicineChina Agricultural University, Beijing 100193, China
| |
Collapse
|
25
|
Mariner-Llicer C, Goig GA, Torres-Puente M, Vashakidze S, Villamayor LM, Saavedra-Cervera B, Mambuque E, Khurtsilava I, Avaliani Z, Rosenthal A, Gabrielian A, Shurgaia M, Shubladze N, García-Basteiro AL, López MG, Comas I. Genetic diversity within diagnostic sputum samples is mirrored in the culture of Mycobacterium tuberculosis across different settings. Nat Commun 2024; 15:7114. [PMID: 39237504 PMCID: PMC11377819 DOI: 10.1038/s41467-024-51266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Culturing and genomic sequencing of Mycobacterium tuberculosis (MTB) from tuberculosis (TB) cases is the basis for many research and clinical applications. The alternative, culture-free sequencing from diagnostic samples, is promising but poses challenges to obtain and analyse the MTB genome. Paradoxically, culture is assumed to impose a diversity bottleneck, which, if true, would entail unexplored consequences. To unravel this paradox we generate high-quality genomes of sputum-culture pairs from two different settings after developing a workflow for sequencing from sputum and a tailored bioinformatics analysis. Careful downstream comparisons reveal sources of sputum-culture incongruences due to false positive/negative variation associated with factors like low input MTB DNA or variable genomic depths. After accounting for these factors, contrary to the bottleneck dogma, we identify a 97% variant agreement within sputum-culture pairs, with a high correlation also in the variants' frequency (0.98). The combined analysis from five different settings and more than 100 available samples shows that our results can be extrapolated to different TB epidemic scenarios, demonstrating that for the cases tested culture accurately mirrors clinical samples.
Collapse
Affiliation(s)
| | - Galo A Goig
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
| | | | - Sergo Vashakidze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- The University of Georgia, Tbilisi, Georgia
| | - Luis M Villamayor
- FISABIO, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, València, Spain
| | - Belén Saavedra-Cervera
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Wellcome Sanger Institute, Hinxton, UK
| | - Edson Mambuque
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Iza Khurtsilava
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- European University, Tbilisi, Georgia
| | - Alex Rosenthal
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Andrei Gabrielian
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Marika Shurgaia
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Natalia Shubladze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- CIBERINFEC, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Barcelona, Spain
| | - Mariana G López
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
| | - Iñaki Comas
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
- CIBERESP, Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain.
| |
Collapse
|
26
|
Qadir M, Khan MT, Khan SA, Akram M, Canseco JO, Faryal R, Wei DQ, Tahseen S. Unveiling the complexity of rifampicin drug susceptibility testing in Mycobacterium tuberculosis: comparative analysis with next-generation sequencing. J Med Microbiol 2024; 73. [PMID: 39229883 DOI: 10.1099/jmm.0.001884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
Introduction. The discordance between phenotypic and molecular methods of rifampicin (RIF) drug susceptibility testing (DST) in Mycobacterium tuberculosis poses a significant challenge, potentially resulting in misdiagnosis and inappropriate treatment.Hypothesis/gap statement. A comparison of RIF phenotypic and molecular methods for DST, including whole genome sequencing (WGS), may provide a better understanding of resistance mechanisms.Aim. This study aims to compare RIF DST in M. tuberculosis using two phenotypic and molecular methods including the GeneXpert RIF Assay (GX) and WGS for better understanding.Methodology. The study evaluated two phenotypic liquid medium methods [Lowenstein-Jensen (LJ) and Mycobacterium Growth Indicator Tube (MGIT)], one targeted molecular method (GX), and one WGS method. Moreover, mutational frequency in ponA1 and ponA2 was also screened in the current and previous RIF resistance M. tuberculosis genomic isolates to find their compensatory role.Results. A total of 25 RIF-resistant isolates, including nine from treatment failures and relapse cases with both discordant and concordant DST results on LJ, MGIT and GX, were subjected to WGS. The phenotypic DST results indicated that 11 isolates (44%) were susceptible on LJ and MGIT but resistant on GX. These isolates exhibited multiple mutations in rpoB, including Thr444>Ala, Leu430>Pro, Leu430>Arg, Asp435>Gly, His445>Asn and Asn438>Lys. Conversely, four isolates that were susceptible on GX and MGIT but resistant on LJ were wild type for rpoB in WGS. However, these isolates possessed several novel mutations in the PonA1 gene, including a 10 nt insertion and two nonsynonymous mutations (Ala394>Ser, Pro631>Ser), as well as one nonsynonymous mutation (Pro780>Arg) in PonA2. The discordance rate of RIF DST is higher on MGIT than on LJ and GX when compared to WGS. These discordances in the Delhi/CAS lineages were primarily associated with failure and relapse cases.Conclusion. The WGS of RIF resistance is relatively expensive, but it may be considered for isolates with discordant DST results on MGIT, LJ and GX to ensure accurate diagnosis and appropriate treatment options.
Collapse
Affiliation(s)
- Mehmood Qadir
- National TB Control Program, National TB Reference Laboratory, Islamabad 44000, Pakistan
- Department of Microbiology, Quaid-i-Azam University, Islamabad 44000, Pakistan
| | - Muhammad Tahir Khan
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, PR China
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan 473006, PR China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 58810, Pakistan
| | - Sajjad Ahmed Khan
- National TB Control Program, National TB Reference Laboratory, Islamabad 44000, Pakistan
| | - Muhammad Akram
- Department of Life Sciences, School of Science, University of Management and Technology, Lahore, Pakistan
| | | | - Rani Faryal
- Department of Microbiology, Quaid-i-Azam University, Islamabad 44000, Pakistan
| | - Dong Qing Wei
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan, 473006, PR China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, PR China
- Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nanshan District, Shenzhen, Guangdong, 518055, PR China
| | - Sabira Tahseen
- National TB Control Program, National TB Reference Laboratory, Islamabad 44000, Pakistan
| |
Collapse
|
27
|
Zhang C, Wu Z, Huang X, Zhao Y, Sun Q, Chen Y, Guo H, Liao Q, Wu H, Chen X, Liang A, Dong W, Yu M, Chen Y, Wei W. A Profile of Drug-Resistant Mutations in Mycobacterium tuberculosis Isolates from Guangdong Province, China. Indian J Microbiol 2024; 64:1044-1056. [PMID: 39282200 PMCID: PMC11399372 DOI: 10.1007/s12088-024-01236-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 09/18/2024] Open
Abstract
Guangdong Province, China's largest economy, has a high incidence of tuberculosis (TB). At present, there are few reports on the distribution, transmission and drug resistance of Mycobacterium tuberculosis (Mtb) strains in this region. In this study, we performed minimum inhibitory concentration testing for 14 anti-TB drugs and whole-genome sequencing of 713 clinical Mtb isolates from 20,662 sputum culture-positive tuberculosis patients registered at 31 tuberculosis drug resistance surveillance sites covering 20 cities in Guangdong Province from 2016 to 2018. Moreover, we evaluated genome-wide associations between mutations and drug resistance, and further investigated the differences in the MICs of mutations. The epidemiology, drug-resistant phenotypes and whole genome sequencing data of 713 clinical Mtb isolates were analyzed, revealing the lineage distribution and drug-resistant gene profiles in Guangdong Province. WGS combined with quantitative MIC measurements identified several novel loci associated with resistance, of which 16 loci were found to be related to resistance to more than one drug. This study analyzed the lineage distribution, prevalence characteristics and resistance-corresponding gene profiles of Mtb isolates in Guangdong province, and provided a theoretical basis for the formulation of tuberculosis prevention and control policy in the province. Supplementary Information The online version contains supplementary material available at 10.1007/s12088-024-01236-3.
Collapse
Affiliation(s)
- Chenchen Zhang
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Zhuhua Wu
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Xinchun Huang
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Yuchuan Zhao
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Qi Sun
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
- Present Address: Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yanmei Chen
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Huixin Guo
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Qinghua Liao
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Huizhong Wu
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Xunxun Chen
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Anqi Liang
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Wenya Dong
- Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, 511443 China
| | - Meiling Yu
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Yuhui Chen
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
| | - Wenjing Wei
- Center for Tuberculosis Control of Guangdong Province, Guangzhou, 510630 China
- College of Basic Medicine and Public Hygiene, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
28
|
Sanchini A, Lanni A, Giannoni F, Mustazzolu A. Exploring diagnostic methods for drug-resistant tuberculosis: A comprehensive overview. Tuberculosis (Edinb) 2024; 148:102522. [PMID: 38850839 DOI: 10.1016/j.tube.2024.102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
Despite available global efforts and funding, Tuberculosis (TB) continues to affect a considerable number of patients worldwide. Policy makers and stakeholders set clear goals to reduce TB incidence and mortality, but the emergence of multidrug-resistant TB (MDR-TB) and extensively drug-resistant TB (XDR-TB) complicate the reach of these goals. Drug-resistance TB needs to be diagnosed rapidly and accurately to effectively treat patients, prevent the transmission of MDR-TB, minimise mortality, reduce treatment costs and avoid unnecessary hospitalisations. In this narrative review, we provide a comprehensive overview of laboratory methods for detecting drug resistance in MTB, focusing on phenotypic, molecular and other drug susceptibility testing (DST) techniques. We found a large variety of methods used, with the BACTEC MGIT 960 being the most common phenotypic DST and the Xpert MTB/RIF being the most common molecular DST. We emphasise the importance of integrating phenotypic and molecular DST to address issues like resistance to new drugs, heteroresistance, mixed infections and low-level resistance mutations. Notably, most of the analysed studies adhered to the outdated definition of XDR-TB and did not consider the pre-XDR definition, thus posing challenges in aligning diagnostic methods with the current landscape of TB resistance.
Collapse
Affiliation(s)
| | - Alessio Lanni
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy.
| | - Federico Giannoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161, Rome, Italy.
| | | |
Collapse
|
29
|
Kontsevaya I, Cabibbe AM, Cirillo DM, DiNardo AR, Frahm N, Gillespie SH, Holtzman D, Meiwes L, Petruccioli E, Reimann M, Ruhwald M, Sabiiti W, Saluzzo F, Tagliani E, Goletti D. Update on the diagnosis of tuberculosis. Clin Microbiol Infect 2024; 30:1115-1122. [PMID: 37490968 DOI: 10.1016/j.cmi.2023.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND Tuberculosis (TB) remains a global public health threat, and the development of rapid and precise diagnostic tools is the key to enabling the early start of treatment, monitoring response to treatment, and preventing the spread of the disease. OBJECTIVES An overview of recent progress in host- and pathogen-based TB diagnostics. SOURCES We conducted a PubMed search of recent relevant articles and guidelines on TB screening and diagnosis. CONTENT An overview of currently used methods and perspectives in the following areas of TB diagnostics is provided: immune-based diagnostics, X-ray, clinical symptoms and scores, cough detection, culture of Mycobacterium tuberculosis and identifying its resistance profile using phenotypic and genotypic methods, including next-generation sequencing, sputum- and non-sputum-based molecular diagnosis of TB and monitoring of response to treatment. IMPLICATIONS A brief overview of the most relevant advances and changes in international guidelines regarding screening and diagnosing TB is provided in this review. It aims at reviewing all relevant areas of diagnostics, including both pathogen- and host-based methods.
Collapse
Affiliation(s)
- Irina Kontsevaya
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany; Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom.
| | | | - Daniela Maria Cirillo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrew R DiNardo
- Global TB Program, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicole Frahm
- Clinical Development, Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | - David Holtzman
- Clinical Development, Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA; Section of Infectious Diseases, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lennard Meiwes
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany
| | - Elisa Petruccioli
- Translational Research Unit, National Institute for Infectious Diseases (INMI) "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Maja Reimann
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany
| | | | - Wilber Sabiiti
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Francesca Saluzzo
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Elisa Tagliani
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases (INMI) "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| |
Collapse
|
30
|
Vasiliauskaitė L, Bakuła Z, Vasiliauskienė E, Bakonytė D, Decewicz P, Dziurzyński M, Proboszcz M, Davidavičienė EV, Nakčerienė B, Krenke R, Kačergius T, Stakėnas P, Jagielski T. Detection of multidrug-resistance in Mycobacterium tuberculosis by phenotype- and molecular-based assays. Ann Clin Microbiol Antimicrob 2024; 23:81. [PMID: 39198827 PMCID: PMC11360294 DOI: 10.1186/s12941-024-00741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND The whole-genome sequencing (WGS) is becoming an increasingly effective tool for rapid and accurate detection of drug resistance in Mycobacterium tuberculosis complex (MTBC). This approach, however, has still been poorly evaluated on strains from Central and Eastern European countries. The purpose of this study was to assess the performance of WGS against conventional drug susceptibility testing (DST) for the detection of multi-drug resistant (MDR) phenotypes among MTBC clinical strains from Poland and Lithuania. METHODS The study included 208 MTBC strains (130 MDR; 78 drug susceptible), recovered from as many tuberculosis patients in Lithuania and Poland between 2018 and 2021. Resistance to rifampicin (RIF) and isoniazid (INH) was assessed by Critical Concentration (CC) and Minimum Inhibitory Concentration (MIC) DST as well as molecular-based techniques, including line-probe assay (LPA) and WGS. The analysis of WGS results was performed using bioinformatic pipeline- and software-based tools. RESULTS The results obtained with the CC DST were more congruent with those by LPA compared to pipeline-based WGS. Software-based tools showed excellent concordance with pipeline-based analysis in prediction of RIF/INH resistance. The RIF-resistant strains demonstrated a relatively homogenous MIC distribution with the mode at the highest tested MIC value. The most frequent RIF-resistance conferring mutation was rpoB S450L. The mode MIC for INH was two-fold higher among double katG and inhA mutants than among single katG mutants. The overall rate of discordant results between all methods was calculated at 5.3%. Three strains had discordant results by both genotypic methods (LPA and pipeline-based WGS), one strain by LPA only, three strains by MIC DST, two strains by both MIC DST and pipeline-based WGS, and the remaining two strains showed discordant results with all three methods, compared to CC DST. CONCLUSIONS Considering MIC DST results, current CCs of the first-line anti-TB drugs might be inappropriately high and may need to be revised. Both molecular methods demonstrated 100% specificity, while pipeline-based WGS had slightly lower sensitivity for RIF and INH than LPA, compared to CC DST.
Collapse
Affiliation(s)
- Laima Vasiliauskaitė
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Centre of Laboratory Medicine, Laboratory of Infectious Diseases and Tuberculosis, Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Zofia Bakuła
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Department of Medical Microbiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Edita Vasiliauskienė
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
- Centre of Laboratory Medicine, Laboratory of Infectious Diseases and Tuberculosis, Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Daiva Bakonytė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Przemysław Decewicz
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | | | - Małgorzata Proboszcz
- Department of Internal Medicine, Pulmonology, and Allergology, Warsaw Medical University, Warsaw, Poland
| | - Edita Valerija Davidavičienė
- Department of Programs and State Tuberculosis Information System, Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Birutė Nakčerienė
- Department of Programs and State Tuberculosis Information System, Vilnius University Hospital Santaros klinikos, Vilnius, Lithuania
| | - Rafał Krenke
- Department of Internal Medicine, Pulmonology, and Allergology, Warsaw Medical University, Warsaw, Poland
| | - Tomas Kačergius
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Petras Stakėnas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Tomasz Jagielski
- Department of Medical Microbiology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
31
|
Deng MZ, Liu Q, Cui SJ, Wang YX, Zhu G, Fu H, Gan M, Xu YY, Cai X, Wang S, Sha W, Zhao GP, Fortune SM, Lyu LD. An additional proofreader contributes to DNA replication fidelity in mycobacteria. Proc Natl Acad Sci U S A 2024; 121:e2322938121. [PMID: 39141351 PMCID: PMC11348249 DOI: 10.1073/pnas.2322938121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
The removal of mis-incorporated nucleotides by proofreading activity ensures DNA replication fidelity. Whereas the ε-exonuclease DnaQ is a well-established proofreader in the model organism Escherichia coli, it has been shown that proofreading in a majority of bacteria relies on the polymerase and histidinol phosphatase (PHP) domain of replicative polymerase, despite the presence of a DnaQ homolog that is structurally and functionally distinct from E. coli DnaQ. However, the biological functions of this type of noncanonical DnaQ remain unclear. Here, we provide independent evidence that noncanonical DnaQ functions as an additional proofreader for mycobacteria. Using the mutation accumulation assay in combination with whole-genome sequencing, we showed that depletion of DnaQ in Mycolicibacterium smegmatis leads to an increased mutation rate, resulting in AT-biased mutagenesis and increased insertions/deletions in the homopolymer tract. Our results showed that mycobacterial DnaQ binds to the β clamp and functions synergistically with the PHP domain proofreader to correct replication errors. Furthermore, the loss of dnaQ results in replication fork dysfunction, leading to attenuated growth and increased mutagenesis on subinhibitory fluoroquinolones potentially due to increased vulnerability to fork collapse. By analyzing the sequence polymorphism of dnaQ in clinical isolates of Mycobacterium tuberculosis (Mtb), we demonstrated that a naturally evolved DnaQ variant prevalent in Mtb lineage 4.3 may enable hypermutability and is associated with drug resistance. These results establish a coproofreading model and suggest a division of labor between DnaQ and PHP domain proofreader. This study also provides real-world evidence that a mutator-driven evolutionary pathway may exist during the adaptation of Mtb.
Collapse
Affiliation(s)
- Ming-Zhi Deng
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
| | - Qingyun Liu
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA02115
| | - Shu-Jun Cui
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai200433, China
| | - Yi-Xin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai200433, China
| | - Guoliang Zhu
- Shanghai Zelixir Biotech Company Ltd., Shanghai200030, China
| | - Han Fu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
- Chinese Academy of Sciences Key Laboratory of Synthetic Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai200032, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Mingyu Gan
- Center for Molecular Medicine, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai201102, China
| | - Yuan-Yuan Xu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
| | - Xia Cai
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd., Shanghai200030, China
| | - Wei Sha
- Shanghai Clinical Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Shanghai200433, China
| | - Guo-Ping Zhao
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai200433, China
- Chinese Academy of Sciences Key Laboratory of Synthetic Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai200032, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA02115
| | - Liang-Dong Lyu
- Key Laboratory of Medical Molecular Virology of the Ministry of Education/Ministry of Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai200032, China
- Shanghai Clinical Research Center for Tuberculosis, Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Shanghai200433, China
| |
Collapse
|
32
|
He G, Zheng Q, Shi J, Wu L, Huang B, Yang Y. Evaluation of WHO catalog of mutations and five WGS analysis tools for drug resistance prediction of Mycobacterium tuberculosis isolates from China. Microbiol Spectr 2024; 12:e0334123. [PMID: 38904370 PMCID: PMC11302272 DOI: 10.1128/spectrum.03341-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
The continuous advancement of molecular diagnostic techniques, particularly whole-genome sequencing (WGS), has greatly facilitated the early diagnosis of drug-resistant tuberculosis patients. Nonetheless, the interpretation of results from various types of mutations in drug-resistant-associated genes has become the primary challenge in the field of molecular drug-resistance diagnostics. In this study, our primary objective is to evaluate the diagnosis accuracy of the World Health Organization (WHO) catalog of mutations and five WGS analysis tools (PhyResSE, Mykrobe, TB Profiler, Gen-TB, and SAM-TB) in drug resistance to 10 anti-Mycobacterium tuberculosis (MTB) drugs. We utilized the data of WGS collected between 2014 and 2017 in Zhejiang Province, consisting of 110 MTB isolates as detailed in our previous study. Based on phenotypic drug susceptibility testing (DST) results using the proportion method on Löwenstein-Jensen medium with antibiotics, we evaluated the predictive accuracy of genotypic DST obtained by these tools. The results revealed that the WHO catalog of mutations and five WGS analysis tools exhibit robust predictive capabilities concerning resistance to isoniazid, rifampicin, ethambutol, streptomycin, amikacin, kanamycin, and capreomycin. Notably, Mykrobe, SAM-TB, and TB Profiler demonstrate the most accurate predictions for resistance to pyrazinamide, prothionamide, and para-aminosalicylic acid, respectively. These findings are poised to significantly guide and influence future clinical treatment strategies and resistance monitoring protocols.IMPORTANCEWhole-genome sequencing (WGS) has the potential for the early diagnosis of drug-resistant tuberculosis. However, the interpretation of mutations of drug-resistant-associated genes represents a significant challenge as the amount and complexity of WGS data. We evaluated the accuracy of the World Health Organization catalog of mutations and five WGS analysis tools in predicting drug resistance to first-line and second-line anti-TB drugs. Our results offer clinicians guidance on selecting appropriate WGS analysis tools for predicting resistance to specific anti-TB drugs.
Collapse
Affiliation(s)
- Guiqing He
- Department of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
- Laboratory of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| | - Qingyong Zheng
- Laboratory of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| | - Jichan Shi
- Department of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| | - Lianpeng Wu
- Department of Clinical Laboratory Medicine, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, Zhejiang, China
| | - Bei Huang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics and Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Yang Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics and Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Animal Science and Technology and College of Veterinary Medicine of Zhejiang A&F University, Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Bahk K, Sung J, Seki M, Kim K, Kim J, Choi H, Whang J, Mitarai S. Pan-lineage Mycobacterium tuberculosis reference genome for enhanced molecular diagnosis. DNA Res 2024; 31:dsae023. [PMID: 39127874 PMCID: PMC11339604 DOI: 10.1093/dnares/dsae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024] Open
Abstract
In Mycobacterium tuberculosis (MTB) control, whole genome sequencing-based molecular drug susceptibility testing (molDST-WGS) has emerged as a pivotal tool. However, the current reliance on a single-strain reference limits molDST-WGS's true potential. To address this, we introduce a new pan-lineage reference genome, 'MtbRf'. We assembled 'unmapped' reads from 3,614 MTB genomes (751 L1; 881 L2; 1,700 L3; and 282 L4) into 35 shared, annotated contigs (54 coding sequences [CDSs]). We constructed MtbRf through: (1) searching for contig homologues among genome database that precipitate results uniquely within Mycobacteria genus; (2) comparing genomes with H37Rv ('lift-over') to define 18 insertions; and (3) filling gaps in H37Rv with insertions. MtbRf adds 1.18% sequences to H37rv, salvaging >60% of previously unmapped reads. Transcriptomics confirmed gene expression of new CDSs. The new variants provided a moderate DST predictive value (AUROC 0.60-0.75). MtbRf thus unveils previously hidden genomic information and lays the foundation for lineage-specific molDST-WGS.
Collapse
Affiliation(s)
- Kunhyung Bahk
- Interdisciplinary Program in Bioinformatics, College of Natural Sciences, Seoul National University, 1, Gwanak-ro, Seoul, 08826, Korea
| | - Joohon Sung
- Interdisciplinary Program in Bioinformatics, College of Natural Sciences, Seoul National University, 1, Gwanak-ro, Seoul, 08826, Korea
- Genome and Health Big Data Laboratory, Graduate School of Public Health, Seoul National University, 1, Gwanak-ro, Seoul, 08826, Korea
- Institute of Health and Environment, Seoul National University, 1, Gwanak-ro, Seoul, 08826, Korea
- Genomic Medicine Institute, Seoul National University College of Medicine, 103, Daehak-ro, Seoul, 03080, Korea
| | - Mitsuko Seki
- Division of Pediatric Dentistry, Department of Human Development and Fostering, Meikai University School of Dentistry, 1-1, Keyakidai, Sakado, Saitama, 350-0283, Japan
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1, Oyaguchi Kami-Cho, Itabashi-Ku, Tokyo, 173-8610, Japan
| | - Kyungjong Kim
- Research and Development Center, The Korean Institute of Tuberculosis, 168-5, Osongsaengmyeong 4-ro, Osong, Cheongju-City, Chungcheongbuk-do, 28158, Korea
- DNA Analysis Division, National Forensic Service, Ministry of the Interior and Safety, 139, Jiyang-ro, Seoul, 08036, Korea
| | - Jina Kim
- Departments of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, 90048, Los Angeles, CA, USA
| | - Hongjo Choi
- Division of Health Policy and Management, Korea University, Seoul, 02841, Korea
| | - Jake Whang
- Research and Development Center, The Korean Institute of Tuberculosis, 168-5, Osongsaengmyeong 4-ro, Osong, Cheongju-City, Chungcheongbuk-do, 28158, Korea
| | - Satoshi Mitarai
- Department of Mycobacterium Reference and Research, Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose, Tokyo, 204-8533Japan
- Department of Basic Mycobacteriology, Graduate School of Biomedical Science, Nagasaki University, Nagasaki, 852-8523Japan
| |
Collapse
|
34
|
Hou Y, Li Y, Tao N, Kong X, Li Y, Liu Y, Li H, Wang Z. Toxin-antitoxin system gene mutations driving Mycobacterium tuberculosis transmission revealed by whole genome sequencing. Front Microbiol 2024; 15:1398886. [PMID: 39144214 PMCID: PMC11322068 DOI: 10.3389/fmicb.2024.1398886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Background The toxin-antitoxin (TA) system plays a vital role in the virulence and pathogenicity of Mycobacterium tuberculosis (M. tuberculosis). However, the regulatory mechanisms and the impact of gene mutations on M. tuberculosis transmission remain poorly understood. Objective To investigate the influence of gene mutations in the toxin-antitoxin system on M. tuberculosis transmission dynamics. Method We performed whole-genome sequencing on the analyzed strains of M. tuberculosis. The genes associated with the toxin-antitoxin system were obtained from the National Center for Biotechnology Information (NCBI) Gene database. Mutations correlating with enhanced transmission within the genes were identified by using random forest, gradient boosting decision tree, and generalized linear mixed models. Results A total of 13,518 M. tuberculosis isolates were analyzed, with 42.29% (n = 5,717) found to be part of genomic clusters. Lineage 4 accounted for the majority of isolates (n = 6488, 48%), followed by lineage 2 (n = 5133, 37.97%). 23 single nucleotide polymorphisms (SNPs) showed a positive correlation with clustering, including vapB1 G34A, vapB24 A76C, vapB2 T171C, mazF2 C85T, mazE2 G104A, vapB31 T112C, relB T226A, vapB11 C54T, mazE5 T344C, vapB14 A29G, parE1 (C103T, C88T), and parD1 C134T. Six SNPs, including vapB6 A29C, vapB31 T112C, parD1 C134T, vapB37 G205C, Rv2653c A80C, and vapB22 C167T, were associated with transmission clades across different countries. Notably, our findings highlighted the positive association of vapB6 A29C, vapB31 T112C, parD1 C134T, vapB37 G205C, vapB19 C188T, and Rv2653c A80C with transmission clades across diverse regions. Furthermore, our analysis identified 32 SNPs that exhibited significant associations with clade size. Conclusion Our study presents potential associations between mutations in genes related to the toxin-antitoxin system and the transmission dynamics of M. tuberculosis. However, it is important to acknowledge the presence of confounding factors and limitations in our study. Further research is required to establish causation and assess the functional significance of these mutations. These findings provide a foundation for future investigations and the formulation of strategies aimed at controlling TB transmission.
Collapse
Affiliation(s)
- Yawei Hou
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifan Li
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Shandong First Medical University (Affiliated Hospital of Shandong Academy of Medical Sciences), Jinan, Shandong, China
| | - Ningning Tao
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xianglong Kong
- Artificial Intelligence Institute Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Yameng Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yao Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huaichen Li
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenguo Wang
- Institute of Chinese Medical Literature and Culture, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
35
|
Che Y, Lu Y, Zhu Y, He T, Li X, Gao J, Gao J, Wang X, Liu Z, Tong F. Surveillance of fluoroquinolones resistance in rifampicin-susceptible tuberculosis in eastern China with whole-genome sequencing-based approach. Front Microbiol 2024; 15:1413618. [PMID: 39050625 PMCID: PMC11266052 DOI: 10.3389/fmicb.2024.1413618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
Background Leveraging well-established DNA-level drug resistance mechanisms, whole-genome sequencing (WGS) has emerged as a valuable methodology for predicting drug resistance. As the most effective second-line anti-tuberculosis (anti-TB) drugs, fluoroquinoloness (FQs) are generally used to treat multidrug-resistant tuberculosis (MDR-TB, defined as being resistant to resistant to rifampicin and isoniazid) or rifampicin-resistant tuberculosis (RR-TB). However, FQs are also commonly used in the management of other bacterial infections. There are few published data on the rates of FQs resistance among rifampicin-susceptible TB. The prevalence of FQs resistance among TB patients who are rifampicin-susceptible has not been studied in Zhejiang Province, China. The goal of this study was to provide a baseline characterization of the prevalence of FQs resistance, particularly among rifampicin-susceptible TB in Zhejiang Province, China. Methods Based on WGS, we have investigated the prevalence of FQs resistance among rifampicin-susceptible TB in Zhejiang Province. All pulmonary TB patients with positive cultures who were identified in Zhejiang area during TB drug resistance surveillance from 2018 to 2019 have enrolled in this population-based retrospective study. Results The rate of FQs resistance was 4.6% (32/698) among TB, 4.0% (27/676) among rifampicin-susceptible TB, and 22.7% (5/22) among RR-TB. According to WGS, strains that differ within 12 single-nucleotide polymorphisms (SNPs) were considered to be transmission of FQ-resistant strains. Specifically, 3.7% (1/27) of FQs resistance was caused by the transmission of FQs-resistant strains among the rifampicin-susceptible TB and 40.7% (11/27) of FQs resistance was identified as hetero-resistance. Conclusion The prevalence of FQs resistance among TB patients who were rifampicin-susceptible was severe in Zhejiang. The emergence of FQs resistance in TB isolates that are rifampicin-susceptible was mainly caused by the selection of drug-resistant strains. In order to prevent the emergence of FQs resistance, the WGS-based surveillance system for TB should be urgently established, and clinical awareness of the responsible use of FQs for respiratory infections should be enhanced.
Collapse
Affiliation(s)
- Yang Che
- Institute of Tuberculosis Prevention and Control, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, China
| | - Yewei Lu
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Yelei Zhu
- The Institute of TB Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Tianfeng He
- Institute of Tuberculosis Prevention and Control, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, China
| | - Xiangchen Li
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Junli Gao
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Junshun Gao
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Hangzhou, China
| | - Xiaomeng Wang
- The Institute of TB Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Zhengwei Liu
- The Institute of TB Control, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Feng Tong
- Institute of Tuberculosis Prevention and Control, Ningbo Municipal Center for Disease Control and Prevention, Ningbo, China
| |
Collapse
|
36
|
Batran RZ, Sabt A, Dziadek J, Kassem AF. Design, synthesis and computational studies of new azaheterocyclic coumarin derivatives as anti- Mycobacterium tuberculosis agents targeting enoyl acyl carrier protein reductase (InhA). RSC Adv 2024; 14:21763-21777. [PMID: 38984262 PMCID: PMC11232110 DOI: 10.1039/d4ra02746a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024] Open
Abstract
In this study, we designed and synthesized a series of coumarin derivatives as antitubercular agents targeting the enoyl acyl carrier protein reductase (InhA) enzyme. Among the synthesized compounds, the tetrazole derivative 4c showed the most potent antitubercular effect with a minimum inhibitory concentration value (MIC) of 15 μg mL-1 against Mtb H37Rv and could also inhibit the growth of the mutant strain (ΔkatG). Compound 4c was able to penetrate Mtb-infected human macrophages and suppress the intracellular growth of tubercle bacilli. Moreover, the target derivative 4c showed a potent inhibitory effect against InhA enzyme with an IC50 value of 0.565 μM, which was superior to the reference InhA inhibitor triclosan. Molecular docking of compound 4c within the InhA active site revealed the importance of the 4-phenylcoumarin ring system and tetrazole moiety for activity. Finally, the physicochemical properties and pharmacokinetic parameters of 4c were investigated.
Collapse
Affiliation(s)
- Rasha Z Batran
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| | - Ahmed Sabt
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences Lodz Poland
| | - Asmaa F Kassem
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki Cairo 12622 Egypt
| |
Collapse
|
37
|
Hiebert M, Sharma MK, Rabb M, Karlowsky L, Bergman K, Soualhine H. Mutations in embB406 Are Associated with Low-Level Ethambutol Resistance in Canadian Mycobacterium tuberculosis Isolates. Antibiotics (Basel) 2024; 13:624. [PMID: 39061306 PMCID: PMC11273804 DOI: 10.3390/antibiotics13070624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
In Mycobacterium tuberculosis, molecular predictions of ethambutol resistance rely primarily on the detection of mutations within embB. However, discordance between embB406 mutations and gold standard phenotypic drug sensitivity testing (DST) questions the significance of embB406 mutations used in molecular DST. This study tabulates embB mutations found in Canadian M. tuberculosis isolates and evaluates the impact of specific mutations on ethambutol resistance. The National Reference Centre for Mycobacteriology culture collection (n = 2796) was screened for isolates with embB mutations. Phenotypic DST was performed on the BACTEC™ MGIT™ 960 at ethambutol concentrations of 2-5 μg/mL. Whole genome sequencing was used for drug resistance predictions, phylogenomics and single nucleotide polymorphism analysis. Detection of resistance-associated embB mutations corresponded to a positive predictive value of 64.3%, negative predictive value of 99.2%, 98.7% specificity, and 73.3% sensitivity compared to phenotypic DST. Two embB406 mutation subtypes (Gly406Asp, Gly406Ala) were found among 16 isolates, of which 12 were sensitive at 5 µg/mL ethambutol with variable resistance between 2-4 µg/mL. A novel frameshift mutation in regulator embR (Gln258fs) was found in nine isolates. Mutations in embB406 were associated with low-level ethambutol resistance undetectable at the recommended critical concentration (5 μg/mL). These novel mutations may exacerbate variability in ethambutol resistance.
Collapse
Affiliation(s)
- Morgan Hiebert
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Meenu K. Sharma
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Melissa Rabb
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
| | - Lisa Karlowsky
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
| | - Kiana Bergman
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
| | - Hafid Soualhine
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.H.); (M.K.S.); (M.R.); (L.K.); (K.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
38
|
Sun Q, Yan J, Long S, Shi Y, Jiang G, Li H, Huang H, Wang G. Apramycin has high in vitro activity against Mycobacterium tuberculosis. J Med Microbiol 2024; 73. [PMID: 38973691 DOI: 10.1099/jmm.0.001854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Introduction. Aminoglycoside antibiotics such as amikacin and kanamycin are important components in the treatment of Mycobacterium tuberculosis (Mtb) infection. However, more and more clinical strains are found to be aminoglycoside antibiotic-resistant. Apramycin is another kind of aminoglycoside antibiotic that is commonly used to treat infections in animals.Hypothesis. Apramycin may have in vitro activity against Mtb.Aim. This study aims to evaluate the efficacy of apramycin against Mtb in vitro and determine its epidemiological cut-off (ECOFF) value.Methodology. One hundred Mtb isolates, including 17 pansusceptible and 83 drug-resistant tuberculosis (DR-TB) strains, were analysed for apramycin resistance using the MIC assay.Results. Apramycin exhibited significant inhibitory activity against Mtb clinical isolates, with an MIC50 of 0.5 μg ml-1 and an MIC90 of 1 μg ml-1. We determined the tentative ECOFF value as 1 µg ml-1 for apramycin. The resistant rates of multidrug-resistant tuberculosis (MDR-TB), pre-extensively drug-resistant (pre-XDR-TB) and extensively drug-resistant tuberculosis (XDR-TB) strains were 12.12 % (4/33), 20.69 % (6/29) and 66.67 % (14/21), respectively. The rrs gene A1401G is associated with apramycin resistance, as well as the cross-resistance between apramycin and other aminoglycosides.Conclusion. Apramycin shows high in vitro activity against the Mtb clinical isolates, especially the MDR-TB clinical isolates. This encouraging discovery calls for more research on the functions of apramycin in vivo and as a possible antibiotic for the treatment of drug-resistant TB.
Collapse
Affiliation(s)
- Qing Sun
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Jun Yan
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Sibo Long
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Yiheng Shi
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| | - Guanglu Jiang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Hao Li
- College of Veterinary Medicine, China Agricultural University, Beijing, PR China
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, PR China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing Chest Hospital, Capital Medical University, Beijing, PR China
| | - Guirong Wang
- Department of Clinical Laboratory, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, PR China
| |
Collapse
|
39
|
Sharma MK, Stobart M, Akochy PM, Adam H, Janella D, Rabb M, Alawa M, Sekirov I, Tyrrell GJ, Soualhine H. Evaluation of Whole Genome Sequencing-Based Predictions of Antimicrobial Resistance to TB First Line Agents: A Lesson from 5 Years of Data. Int J Mol Sci 2024; 25:6245. [PMID: 38892433 PMCID: PMC11172968 DOI: 10.3390/ijms25116245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Phenotypic susceptibility testing of the Mycobacterium tuberculosis complex (MTBC) isolate requires culture growth, which can delay rapid detection of resistant cases. Whole genome sequencing (WGS) and data analysis pipelines can assist in predicting resistance to antimicrobials used in the treatment of tuberculosis (TB). This study compared phenotypic susceptibility testing results and WGS-based predictions of antimicrobial resistance (AMR) to four first-line antimicrobials-isoniazid, rifampin, ethambutol, and pyrazinamide-for MTBC isolates tested between the years 2018-2022. For this 5-year retrospective analysis, the WGS sensitivity for predicting resistance for isoniazid, rifampin, ethambutol, and pyrazinamide using Mykrobe was 86.7%, 100.0%, 100.0%, and 47.8%, respectively, and the specificity was 99.4%, 99.5%, 98.7%, and 99.9%, respectively. The predictive values improved slightly using Mykrobe corrections applied using TB Profiler, i.e., the WGS sensitivity for isoniazid, rifampin, ethambutol, and pyrazinamide was 92.31%, 100%, 100%, and 57.78%, respectively, and the specificity was 99.63%. 99.45%, 98.93%, and 99.93%, respectively. The utilization of WGS-based testing addresses concerns regarding test turnaround time and enables analysis for MTBC member identification, antimicrobial resistance prediction, detection of mixed cultures, and strain genotyping, all through a single laboratory test. WGS enables rapid resistance detection compared to traditional phenotypic susceptibility testing methods using the WHO TB mutation catalog, providing an insight into lesser-known mutations, which should be added to prediction databases as high-confidence mutations are recognized. The WGS-based methods can support TB elimination efforts in Canada and globally by ensuring the early start of appropriate treatment, rapidly limiting the spread of TB outbreaks.
Collapse
Affiliation(s)
- Meenu Kaushal Sharma
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada (M.S.)
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Michael Stobart
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada (M.S.)
| | - Pierre-Marie Akochy
- Laboratoire de Santé Publique du Québec-Institut National de Santé Publique du Québec, Sainte-Anne-de-Bellevue, QC H9X 3R5, Canada
| | - Heather Adam
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Diagnostic Services, Shared Health, Winnipeg, MB R3C 3H8, Canada
| | - Debra Janella
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada (M.S.)
| | - Melissa Rabb
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada (M.S.)
| | - Mohey Alawa
- Regina Qu’Appelle Health Region, Regina, SK S4T 1A5, Canada;
| | - Inna Sekirov
- Public Health Laboratory, B.C. Centre for Disease Control, Vancouver, BC V5Z 4R4, Canada;
| | - Gregory J. Tyrrell
- Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2J2, Canada
- Alberta Precision Laboratories Public Health, Edmonton, AB T6G 2J2, Canada
| | - Hafid Soualhine
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada (M.S.)
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| |
Collapse
|
40
|
He G, Zheng Q, Wu J, Wu L, Geng Z, Jiang G, Huang H, Jiang X, Yu X. Discordant results between Xpert MTB/RIF assay and Bactec MGIT 960 culture system regarding the detection of rifampin-resistant Mycobacterium tuberculosis isolates in Wenzhou, China. Microbiol Spectr 2024; 12:e0385923. [PMID: 38738892 PMCID: PMC11237732 DOI: 10.1128/spectrum.03859-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
This study aimed to assess the possible causes of discordant results between Xpert MTB/RIF (Xpert) and Bactec MGIT 960 Culture System (MGIT960) regarding rifampicin (RIF) susceptibility in Mycobacterium tuberculosis. Patients with previous RIF-resistant tuberculosis who were admitted to Wenzhou Central Hospital from January 2020 to December 2022 were enrolled. The isolates obtained from these patients were subjected to RIF susceptibility tests using Xpert and MGIT960, and the minimum inhibitory concentration (MIC) of RIF was determined by the MYCOTB MIC plate test. Additionally, molecular docking and molecular dynamics (MD) simulations were performed to evaluate the binding efficacy of rpoB and RIF based on rpoB mutations detected in the isolates with discordant RIF susceptibility results. A total of 28 isolates with discordant RIF susceptibility test results were detected, 15 of them were RIF susceptible with MICs ≤ 0.5 µg/mL. Twelve out of 15 isolates contained borderline RIF resistance-associated mutations [L430P (n = 6), H445N (n = 6)], 1 isolate had D435Y and Q429H double mutation, and the remaining 2 isolates had a silent (Q432Q) mutation. Compared with the affinity of RIF toward the wild type (WT) (-45.83 kcal/mol) by MD, its affinity toward L452P (-55.52 kcal/mol), D435Y (-47.39 kcal/mol), L430P (approximately -69.72 kcal/mol), H445N (-49.53 kcal/mol), and Q429H (-55.67 kcal/mol) increased. Borderline RIF resistance-associated mutations were the main cause for the discordant RIF susceptibility results between Xpert and MGIT960, and the mechanisms of the resistance need further investigated.IMPORTANCEThis study is aimed at assessing discordant results between Xpert MTB/RIF (Xpert) assay and Bactec MGIT 960 Culture System (MGIT960) regarding the detection of rifampicin (RIF)-resistant Mycobacterium tuberculosis isolates in Wenzhou, China. The discordant results of RIF between these two assays were mainly caused by borderline RIF resistance-associated mutations, subsequently by silent mutations of rpoB. Borderline RIF resistance- associated mutations detected in our study were demonstrated to not be affected by the affinity of rpoB and RIF by molecular dynamics, and the mechanism of resistance was needed to be clarified. For the discordant results of RIF by Xpert and MGIT960 that occurred, rpoB DNA sequencing was recommended to investigate its association with resistance to RIF.
Collapse
Affiliation(s)
- Guiqing He
- Department of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
- Laboratory of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Qingyong Zheng
- Laboratory of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Jing Wu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Lianpeng Wu
- Department of Clinical Laboratory Medicine, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Zhi Geng
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Guanglu Jiang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hairong Huang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Xiangao Jiang
- Department of Infectious Diseases, Wenzhou Central Hospital, The Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Xia Yu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Kolte B, Nübel U. Genetic determinants of resistance to antimicrobial therapeutics are rare in publicly available Clostridioides difficile genome sequences. J Antimicrob Chemother 2024; 79:1320-1328. [PMID: 38598696 PMCID: PMC11144481 DOI: 10.1093/jac/dkae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
OBJECTIVES To determine the frequencies and clonal distributions of putative genetic determinants of resistance to antimicrobials applied for treatment of Clostridioides difficile infection (CDI), as documented in the genomic record. METHODS We scanned 26 557 C. difficile genome sequences publicly available from the EnteroBase platform for plasmids, point mutations and gene truncations previously reported to reduce susceptibility to vancomycin, fidaxomicin or metronidazole, respectively. We measured the antimicrobial susceptibility of 143 selected C. difficile isolates. RESULTS The frequency of mutations causing reduced susceptibility to vancomycin and metronidazole, respectively, increased strongly after 2000, peaking at up to 52% of all sequenced C. difficile genomes. However, both mutations declined sharply more recently, reflecting major changes in CDI epidemiology. We detected mutations associated with fidaxomicin resistance in several major genotypes, but found no evidence of international spread of resistant clones. The pCD-METRO plasmid, conferring metronidazole resistance, was detected in a single previously unreported C. difficile isolate, recovered from a hospital patient in Germany in 2008. The pX18-498 plasmid, putatively associated with decreased vancomycin susceptibility, was confined to related, recent isolates from the USA. Phenotype measurements confirmed that most of those genetic features were useful predictors of antibiotic susceptibility, even though ranges of MICs typically overlapped among isolates with and without specific mutations. CONCLUSIONS Genomic data suggested that resistance to therapeutic antimicrobial drugs is rare in C. difficile. Public antimicrobial resistance marker databases were not equipped to detect most of the genetic determinants relevant to antibiotic therapy of CDI.
Collapse
Affiliation(s)
- Baban Kolte
- Leibniz Institute DSMZ—German Collection of Microorganisms and Cell Cultures, Microbial Genome Research, Inhoffenstr. 7B, 38124, Braunschweig, Germany
- Technical University Braunschweig, Institute of Microbiology, Braunschweig, Germany
| | - Ulrich Nübel
- Leibniz Institute DSMZ—German Collection of Microorganisms and Cell Cultures, Microbial Genome Research, Inhoffenstr. 7B, 38124, Braunschweig, Germany
- Technical University Braunschweig, Institute of Microbiology, Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Braunschweig-Hannover, Braunschweig, Germany
| |
Collapse
|
42
|
Cloutier Charette W, Rabodoarivelo MS, Point F, Knoblauch AM, Andrianomanana FR, Hall MB, Iqbal Z, Supply P, Martin A, Rakotosamimanana N, Grandjean Lapierre S. Concordance of targeted and whole genome sequencing for Mycobacterium tuberculosis genotypic drug susceptibility testing. Diagn Microbiol Infect Dis 2024; 109:116249. [PMID: 38537504 DOI: 10.1016/j.diagmicrobio.2024.116249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/30/2024]
Abstract
Targeted Next Generation Sequencing (tNGS) and Whole Genome Sequencing (WGS) are increasingly used for genotypic drug susceptibility testing (gDST) of Mycobacterium tuberculosis. Thirty-two multi-drugs resistant and 40 drug susceptible isolates from Madagascar were tested with Deeplex® Myc-TB and WGS using the Mykrobe analysis pipeline. Sixty-four of 72 (89 %) yielded concordant categorical gDST results for drugs tested by both assays. Mykrobe didn't detect pncA K96T, pncA Q141P, pncA H51P, pncA H82R, rrs C517T and rpsL K43R mutations, which were identified as minority variants in corresponding isolates by tNGS. One discrepancy (rrs C517T) was associated with insufficient sequencing depth on WGS. Deeplex® Myc-TB didn't detect inhA G-154A which isn't covered by the assay's amplification targets. Despite those targets being included in the Deeplex® Myc-TB assay, a pncA T47A and a deletion in gid were not identified in one isolate respectively. The evaluated WGS and tNGS gDST assays show high but imperfect concordance.
Collapse
Affiliation(s)
- William Cloutier Charette
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montréal, Québec, Canada; Immunopathology Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Marie-Sylvianne Rabodoarivelo
- Mycobacteriology Unit, Institut Pasteur de Madagascar, Antananarivo, Madagascar; Departamento de Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, Spain
| | - Floriane Point
- Immunopathology Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | - Astrid M Knoblauch
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | | | - Michael B Hall
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Zamin Iqbal
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | - Philip Supply
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Anandi Martin
- Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Belgium
| | | | - Simon Grandjean Lapierre
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montréal, Québec, Canada; Immunopathology Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada; Mycobacteriology Unit, Institut Pasteur de Madagascar, Antananarivo, Madagascar.
| |
Collapse
|
43
|
Duffey M, Shafer RW, Timm J, Burrows JN, Fotouhi N, Cockett M, Leroy D. Combating antimicrobial resistance in malaria, HIV and tuberculosis. Nat Rev Drug Discov 2024; 23:461-479. [PMID: 38750260 DOI: 10.1038/s41573-024-00933-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 06/07/2024]
Abstract
Antimicrobial resistance poses a significant threat to the sustainability of effective treatments against the three most prevalent infectious diseases: malaria, human immunodeficiency virus (HIV) infection and tuberculosis. Therefore, there is an urgent need to develop novel drugs and treatment protocols capable of reducing the emergence of resistance and combating it when it does occur. In this Review, we present an overview of the status and underlying molecular mechanisms of drug resistance in these three diseases. We also discuss current strategies to address resistance during the research and development of next-generation therapies. These strategies vary depending on the infectious agent and the array of resistance mechanisms involved. Furthermore, we explore the potential for cross-fertilization of knowledge and technology among these diseases to create innovative approaches for minimizing drug resistance and advancing the discovery and development of new anti-infective treatments. In conclusion, we advocate for the implementation of well-defined strategies to effectively mitigate and manage resistance in all interventions against infectious diseases.
Collapse
Affiliation(s)
- Maëlle Duffey
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland
- The Global Antibiotic Research & Development Partnership, Geneva, Switzerland
| | - Robert W Shafer
- Department of Medicine/Infectious Diseases, Stanford University, Palo Alto, CA, USA
| | | | - Jeremy N Burrows
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland
| | | | | | - Didier Leroy
- Medicines for Malaria Venture (MMV), R&D Department/Drug Discovery, ICC, Geneva, Switzerland.
| |
Collapse
|
44
|
Billows N, Phelan J, Xia D, Peng Y, Clark TG, Chang YM. Large-scale statistical analysis of Mycobacterium tuberculosis genome sequences identifies compensatory mutations associated with multi-drug resistance. Sci Rep 2024; 14:12312. [PMID: 38811658 PMCID: PMC11137121 DOI: 10.1038/s41598-024-62946-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, has a significant impact on global health worldwide. The development of multi-drug resistant strains that are resistant to the first-line drugs isoniazid and rifampicin threatens public health security. Rifampicin and isoniazid resistance are largely underpinned by mutations in rpoB and katG respectively and are associated with fitness costs. Compensatory mutations are considered to alleviate these fitness costs and have been observed in rpoC/rpoA (rifampicin) and oxyR'-ahpC (isoniazid). We developed a framework (CompMut-TB) to detect compensatory mutations from whole genome sequences from a large dataset comprised of 18,396 M. tuberculosis samples. We performed association analysis (Fisher's exact tests) to identify pairs of mutations that are associated with drug-resistance, followed by mediation analysis to identify complementary or full mediators of drug-resistance. The analyses revealed several potential mutations in rpoC (N = 47), rpoA (N = 4), and oxyR'-ahpC (N = 7) that were considered either 'highly likely' or 'likely' to confer compensatory effects on drug-resistance, including mutations that have previously been reported and validated. Overall, we have developed the CompMut-TB framework which can assist with identifying compensatory mutations which is important for more precise genome-based profiling of drug-resistant TB strains and to further understanding of the evolutionary mechanisms that underpin drug-resistance.
Collapse
Affiliation(s)
- Nina Billows
- Royal Veterinary College, University of London, London, UK.
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | - Jody Phelan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Dong Xia
- Royal Veterinary College, University of London, London, UK
| | | | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Yu-Mei Chang
- Royal Veterinary College, University of London, London, UK
| |
Collapse
|
45
|
Singh M, Dhanwal A, Verma A, Augustin L, Kumari N, Chakraborti S, Agarwal N, Sriram D, Dey RJ. Discovery of potent antimycobacterial agents targeting lumazine synthase (RibH) of Mycobacterium tuberculosis. Sci Rep 2024; 14:12170. [PMID: 38806590 PMCID: PMC11133327 DOI: 10.1038/s41598-024-63051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 05/24/2024] [Indexed: 05/30/2024] Open
Abstract
Tuberculosis (TB) continues to be a global health crisis, necessitating urgent interventions to address drug resistance and improve treatment efficacy. In this study, we validate lumazine synthase (RibH), a vital enzyme in the riboflavin biosynthetic pathway, as a potential drug target against Mycobacterium tuberculosis (M. tb) using a CRISPRi-based conditional gene knockdown strategy. We employ a high-throughput molecular docking approach to screen ~ 600,000 compounds targeting RibH. Through in vitro screening of 55 shortlisted compounds, we discover 3 compounds that exhibit potent antimycobacterial activity. These compounds also reduce intracellular burden of M. tb during macrophage infection and prevent the resuscitation of the nutrient-starved persister bacteria. Moreover, these three compounds enhance the bactericidal effect of first-line anti-TB drugs, isoniazid and rifampicin. Corroborating with the in silico predicted high docking scores along with favourable ADME and toxicity profiles, all three compounds demonstrate binding affinity towards purified lumazine synthase enzyme in vitro, in addition these compounds exhibit riboflavin displacement in an in vitro assay with purified lumazine synthase indicative of specificity of these compounds to the active site. Further, treatment of M. tb with these compounds indicate reduced production of flavin adenine dinucleotide (FAD), the ultimate end product of the riboflavin biosynthetic pathway suggesting the action of these drugs on riboflavin biosynthesis. These compounds also show acceptable safety profile in mammalian cells, with a high selective index. Hence, our study validates RibH as an important drug target against M. tb and identifies potent antimycobacterial agents.
Collapse
Affiliation(s)
- Monica Singh
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India
| | - Anannya Dhanwal
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India
| | - Arpita Verma
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India
| | - Linus Augustin
- Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Niti Kumari
- National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, 500032, India
| | - Soumyananda Chakraborti
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India
- National Institute of Malaria Research, Indian Council of Medical Research (ICMR), New Delhi, 110077, India
| | - Nisheeth Agarwal
- Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India
| | - Ruchi Jain Dey
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
46
|
Guo J, Han Y, Zhang X, Lin F, Chen L, Feng X. Risk factors of adult isoniazid-resistant and rifampicin-susceptible tuberculosis in Nanjing, 2019-2021. BMC Infect Dis 2024; 24:511. [PMID: 38773443 PMCID: PMC11110291 DOI: 10.1186/s12879-024-09404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
INTRODUCTION This study aimed to analyze the risk factors associated with isoniazid-resistant and rifampicin-susceptible tuberculosis (Hr-TB) in adults. METHOD The clinical data of 1,844 adult inpatients diagnosed with culture-positive pulmonary tuberculosis (PTB) in Nanjing Second Hospital from January 2019 and December 2021 were collected. All culture positive strain from the patient specimens underwent drug susceptibility testing (DST). Among them, 166 patients with Hr-TB were categorized as the Hr-TB group, while the remaining 1,678 patients were classified as having drug-susceptible tuberculosis (DS-TB). Hierarchical logistic regression was employed for multivariate analysis to identify variables associated with Hr-TB. RESULTS Multivariate logistic regression analysis revealed that individuals with diabetes mellitus (DM) (OR 1.472, 95% CI 1.037-2.088, p = 0.030) and a history of previous tuberculosis treatment (OR 2.913, 95% CI 1.971-4.306, p = 0.000) were at higher risk of developing adult Hr-TB, with this risk being more pronounced in male patients. Within the cohort, 1,640 patients were newly treated, and among them, DM (OR 1.662, 95% CI 1.123-2.461, p = 0.011) was identified as risk factors for Hr-TB. CONCLUSIONS Diabetes mellitus is a risk factor for Hr-TB in adults, and the contribution of diabetes as a risk factor was more pronounced in the newly treatment or male subgroup. And previous TB treatment history is also a risk factor for Hr-TB in adults.
Collapse
Affiliation(s)
- Jing Guo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, PR China
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China
| | - Yan Han
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, PR China
| | - Feishen Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, PR China
| | - Liangyu Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, Jiangsu Province, 210008, PR China
| | - Xuebing Feng
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 211132, China.
| |
Collapse
|
47
|
Liu S, Wen H, Li F, Xue X, Sun X, Li F, Hu R, Xi H, Boccellato F, Meyer TF, Mi Y, Zheng P. Revealing the pathogenesis of gastric intestinal metaplasia based on the mucosoid air-liquid interface. J Transl Med 2024; 22:468. [PMID: 38760813 PMCID: PMC11101349 DOI: 10.1186/s12967-024-05276-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Gastric intestinal metaplasia (GIM) is an essential precancerous lesion. Although the reversal of GIM is challenging, it potentially brings a state-to-art strategy for gastric cancer therapeutics (GC). The lack of the appropriate in vitro model limits studies of GIM pathogenesis, which is the issue this work aims to address for further studies. METHOD The air-liquid interface (ALI) model was adopted for the long-term culture of GIM cells in the present work. This study conducted Immunofluorescence (IF), quantitative real-time polymerase chain reaction (qRT-PCR), transcriptomic sequencing, and mucoproteomic sequencing (MS) techniques to identify the pathways for differential expressed genes (DEGs) enrichment among different groups, furthermore, to verify novel biomarkers of GIM cells. RESULT Our study suggests that GIM-ALI model is analog to the innate GIM cells, which thus can be used for mucus collection and drug screening. We found genes MUC17, CDA, TRIM15, TBX3, FLVCR2, ONECUT2, ACY3, NMUR2, and MAL2 were highly expressed in GIM cells, while GLDN, SLC5A5, MAL, and MALAT1 showed down-regulated, which can be used as potential biomarkers for GIM cells. In parallel, these genes that highly expressed in GIM samples were mainly involved in cancer-related pathways, such as the MAPK signal pathway and oxidative phosphorylation signal pathway. CONCLUSION The ALI model is validated for the first time for the in vitro study of GIM. GIM-ALI model is a novel in vitro model that can mimic the tissue micro-environment in GIM patients and further provide an avenue for studying the characteristics of GIM mucus. Our study identified new markers of GIM as well as pathways associated with GIM, which provides outstanding insight for exploring GIM pathogenesis and potentially other related conditions.
Collapse
Affiliation(s)
- Simeng Liu
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117, Berlin, Germany
| | - Huijuan Wen
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
| | - Fazhan Li
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
| | - Xia Xue
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
| | - Xiangdong Sun
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
| | - Fuhao Li
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
| | - Ruoyu Hu
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 453000, China
| | - Huayuan Xi
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 453000, China
| | - Francesco Boccellato
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117, Berlin, Germany
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, 11743, UK
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117, Berlin, Germany
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Christian Albrecht University of Kiel and University Hospital Schleswig-Holstein - Campus Kiel, Rosalind-Franklin- Straße 12, 24105, Kiel, Germany
| | - Yang Mi
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China.
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3, Kangfuqian Street, Erqi District, Zhengzhou, Henan, 450002, China.
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 453000, China.
| |
Collapse
|
48
|
Tagami Y, Horita N, Kaneko M, Muraoka S, Fukuda N, Izawa A, Kaneko A, Somekawa K, Kamimaki C, Matsumoto H, Tanaka K, Murohashi K, Aoki A, Fujii H, Watanabe K, Hara Y, Kobayashi N, Kaneko T. Whole-Genome Sequencing Predicting Phenotypic Antitubercular Drug Resistance: Meta-analysis. J Infect Dis 2024; 229:1481-1492. [PMID: 37946558 DOI: 10.1093/infdis/jiad480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 10/06/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND For simultaneous prediction of phenotypic drug susceptibility test (pDST) for multiple antituberculosis drugs, the whole genome sequencing (WGS) data can be analyzed using either a catalog-based approach, wherein 1 causative mutation suggests resistance, (eg, World Health Organization catalog) or noncatalog-based approach using complicated algorithm (eg, TB-profiler, machine learning). The aim was to estimate the predictive ability of WGS-based tests with pDST as the reference, and to compare the 2 approaches. METHODS Following a systematic literature search, the diagnostic test accuracies for 14 drugs were pooled using a random-effect bivariate model. RESULTS Of 779 articles, 44 with 16 821 specimens for meta-analysis and 13 not for meta-analysis were included. The areas under summary receiver operating characteristic curve suggested test accuracy was excellent (0.97-1.00) for 2 drugs (isoniazid 0.975, rifampicin 0.975), very good (0.93-0.97) for 8 drugs (pyrazinamide 0.946, streptomycin 0.952, amikacin 0.968, kanamycin 0.963, capreomycin 0.965, para-aminosalicylic acid 0.959, levofloxacin 0.960, ofloxacin 0.958), and good (0.75-0.93) for 4 drugs (ethambutol 0.926, moxifloxacin 0.896, ethionamide 0.878, prothionamide 0.908). The noncatalog-based and catalog-based approaches had similar ability for all drugs. CONCLUSIONS WGS accurately identifies isoniazid and rifampicin resistance. For most drugs, positive WGS results reliably predict pDST positive. The 2 approaches had similar ability. CLINICAL TRIALS REGISTRATION UMIN-ID UMIN000049276.
Collapse
Affiliation(s)
- Yoichi Tagami
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuyuki Horita
- Chemotherapy Center, Yokohama City University Hospital, Yokohama, Japan
| | - Megumi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Suguru Muraoka
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuhiko Fukuda
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ami Izawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ayami Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kohei Somekawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Chisato Kamimaki
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromi Matsumoto
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Katsushi Tanaka
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kota Murohashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ayako Aoki
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroaki Fujii
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keisuke Watanabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
49
|
Bahuaud O, Genestet C, Hodille E, Vallée M, Testard Q, Tataï C, Saison J, Rasigade JP, Lina G, Ader F, Dumitrescu O. Rapid resistance detection is reliable for prompt adaptation of isoniazid resistant tuberculosis management. Heliyon 2024; 10:e29932. [PMID: 38726207 PMCID: PMC11078763 DOI: 10.1016/j.heliyon.2024.e29932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
Objectives Appropriate tuberculosis (TB) management requires anti-TB drugs resistance detection. We assessed the performance of rapid resistance detection assays and their impact on treatment adaptation, focusing on isoniazid resistant (Hr) TB. Methods From 2016 to 2022, all TB cases enrolled in 3 hospitals were reviewed for phenotypic drug susceptibility testing (p-DST) and genotypic DST (g-DST) performed by rapid molecular testing, and next generation sequencing (NGS). Clinical characteristics, treatment and outcome were collected for Hr-TB patients. The concordance between g-DST and p-DST results, and delay between treatment initiation and results of g-DST and p-DST were respectively recorded to assess the contribution of DST results on Hr-TB management. Results Among 654 TB cases enrolled, 29 were Hr-TB. Concordance between g-DST by rapid molecular methods and p-DST was 76.9 %, whilst concordance between NGS-based g-DST and p-DST was 98.7 %. Rapid resistance detection significantly fastened Hr-TB treatment adaptation (median delay between g-DST results and treatment modification was 6 days). It consisted in fluoroquinolone implementation for 17/23 patients; outcome was favourable except for 2 patients who died before DST reporting. Conclusion Rapid resistance detection fastened treatment adaptation. Also, NGS-based g-DST showed almost perfect concordance with p-DST, thus providing rapid and safe culture-free DST alternative.
Collapse
Affiliation(s)
- Olivier Bahuaud
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Service des Maladies Infectieuses et Tropicales, Lyon, France
| | - Charlotte Genestet
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Elisabeth Hodille
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Maxime Vallée
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Quentin Testard
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Caroline Tataï
- Centre de Lutte Anti Tuberculeuse, Bourg-en-Bresse, France
| | - Julien Saison
- Infectious Diseases Department, Valence Hospital Center, Valence, France
- Clinical Research Unit, Valence Hospital Center, Valence, France
| | - Jean-Philippe Rasigade
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Gérard Lina
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
- Université Lyon 1, Facultés de Médecine et de Pharmacie de Lyon, Lyon, France
| | - Florence Ader
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Service des Maladies Infectieuses et Tropicales, Lyon, France
| | - Oana Dumitrescu
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
- Université Lyon 1, Facultés de Médecine et de Pharmacie de Lyon, Lyon, France
| | - Lyon TB study group
- CIRI - Centre International de Recherche en Infectiologie, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon-1, Inserm U1111, CNRS UMR5308, Lyon, France
- Hospices Civils de Lyon, Service des Maladies Infectieuses et Tropicales, Lyon, France
- Hospices Civils de Lyon, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
- Centre de Lutte Anti Tuberculeuse, Bourg-en-Bresse, France
- Infectious Diseases Department, Valence Hospital Center, Valence, France
- Clinical Research Unit, Valence Hospital Center, Valence, France
- Université Lyon 1, Facultés de Médecine et de Pharmacie de Lyon, Lyon, France
| |
Collapse
|
50
|
Poonawala H, Zhang Y, Kuchibhotla S, Green AG, Cirillo DM, Di Marco F, Spitlaeri A, Miotto P, Farhat MR. Transcriptomic responses to antibiotic exposure in Mycobacterium tuberculosis. Antimicrob Agents Chemother 2024; 68:e0118523. [PMID: 38587412 PMCID: PMC11064486 DOI: 10.1128/aac.01185-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 03/06/2024] [Indexed: 04/09/2024] Open
Abstract
Transcriptional responses in bacteria following antibiotic exposure offer insights into antibiotic mechanism of action, bacterial responses, and characterization of antimicrobial resistance. We aimed to define the transcriptional antibiotic response (TAR) in Mycobacterium tuberculosis (Mtb) isolates for clinically relevant drugs by pooling and analyzing Mtb microarray and RNA-seq data sets. We generated 99 antibiotic transcription profiles across 17 antibiotics, with 76% of profiles generated using 3-24 hours of antibiotic exposure and 49% within one doubling of the WHO antibiotic critical concentration. TAR genes were time-dependent, and largely specific to the antibiotic mechanism of action. TAR signatures performed well at predicting antibiotic exposure, with the area under the receiver operating curve (AUC) ranging from 0.84-1.00 (TAR <6 hours of antibiotic exposure) and 0.76-1.00 (>6 hours of antibiotic exposure) for upregulated genes and 0.57-0.90 and 0.87-1.00, respectfully, for downregulated genes. This work desmonstrates that transcriptomics allows for the assessment of antibiotic activity in Mtb within 6 hours of exposure.
Collapse
Affiliation(s)
- Husain Poonawala
- Department of Medicine and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Medicine and Department of Anatomic and Clinical Pathology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Yu Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Anna G. Green
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniela Maria Cirillo
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Di Marco
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Spitlaeri
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Paolo Miotto
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maha R. Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|