1
|
Dartois VA, Mizrahi V, Savic RM, Silverman JA, Hermann D, Barry CE. Strategies for shortening tuberculosis therapy. Nat Med 2025:10.1038/s41591-025-03742-3. [PMID: 40514466 DOI: 10.1038/s41591-025-03742-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 04/25/2025] [Indexed: 06/16/2025]
Abstract
In the absence of effective patient-stratification approaches, tuberculosis (TB) treatment relies on overtreating most patients to ensure high cure rates. Shortening treatment duration without compromising efficacy is therefore high on the agenda of the global TB community. While new and better drugs are certainly needed, we argue that innovative but rational treatment strategies, using both new and existing therapies, will help achieve this goal. There is growing recognition that patient stratification, based on host and pathogen factors, is key to delivering the right drug regimen for the right duration. In this Perspective, we review the current knowledge on the heterogeneity of TB disease and propose approaches to optimize treatment duration in distinct patient groups, taking into consideration the realities of TB control globally. We emphasize key insights that improve the understanding of bacterial vulnerabilities in patients with easy-to-treat and hard-to-treat TB, helping to reduce diagnostic uncertainties. We explore how the TB research community can integrate disease biology, pathology and symptoms, to rethink therapeutic strategies and reduce TB treatment duration.
Collapse
Affiliation(s)
- Véronique A Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, NJ, USA.
| | - Valerie Mizrahi
- Molecular Mycobacteriology Research Unit, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Radojka M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Alberts ME, Kurtz MP, Müh U, Bernardi JP, Bollinger KW, Dobrila HA, Duncan L, Laster HM, Orea AJ, Pannullo AG, Rivera-Rosado JG, Torres FV, Ellermeier CD, Weiss DS. Analysis of Essential Genes in Clostridioides difficile by CRISPRi and Tn-seq. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.06.04.657922. [PMID: 40502013 PMCID: PMC12157513 DOI: 10.1101/2025.06.04.657922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
Essential genes are interesting in their own right and as potential antibiotic targets. To date, only one report has identified essential genes on a genome-wide scale in Clostridioides difficile, a problematic pathogen for which treatment options are limited. That foundational study used large-scale transposon mutagenesis to identify 404 protein-encoding genes as likely to be essential for vegetative growth of the epidemic strain R20291. Here, we revisit the essential genes of strain R20291 using a combination of CRISPR interference (CRISPRi) and transposon-sequencing (Tn-seq). First, we targeted 181 of the 404 putatively essential genes with CRISPRi. We confirmed essentiality for >90% of the targeted genes and observed morphological defects for >80% of them. Second, we conducted a new Tn-seq analysis, which identified 346 genes as essential, of which 283 are in common with the previous report and might be considered a provisional essential gene set that minimizes false positives. We compare the list of essential genes to those of other bacteria, especially Bacillus subtilis, highlighting some noteworthy differences. Finally, we used fusions to red fluorescent protein (RFP) to identify 18 putative new cell division proteins, three of which are conserved in Bacillota but of largely unknown function. Collectively, our findings provide new tools and insights that advance our understanding of C. difficile.
Collapse
Affiliation(s)
- Maia E. Alberts
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Present address: Department of Anesthesiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Micaila P. Kurtz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ute Müh
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jonathon P. Bernardi
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kevin W. Bollinger
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Horia A. Dobrila
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Present address: Departments of Medicine and Medical Microbiology & Immunology, University of Wisconsin, Madison, WI, USA
| | | | - Hannah M. Laster
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Andres J. Orea
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Present address: Department of Pharmacology, University of California, Davis, CA, USA
| | - Anthony G. Pannullo
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Present address: JMI Laboratories, North Liberty, IA, USA
| | - Juan G. Rivera-Rosado
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Facundo V. Torres
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Present address: Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Craig D. Ellermeier
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - David S. Weiss
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
3
|
Lepori I, Jackson K, Liu Z, Chordia MD, Wong M, Rivera SL, Roncetti M, Poliseno L, Freundlich JS, Pires MM, Siegrist MS. The Mycomembrane Differentially and Heterogeneously Restricts Antibiotic Permeation. ACS Infect Dis 2025. [PMID: 40471697 DOI: 10.1021/acsinfecdis.4c01062] [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: 06/11/2025]
Abstract
The recalcitrance of Mycobacterium tuberculosis to antibiotic treatment has been broadly attributed to the impermeability of the organism's outer mycomembrane. However, the studies that support this inference have been indirect or reliant on bulk population measurements. We previously developed the Peptidoglycan Accessibility Click-Mediated AssessmeNt (PAC-MAN) method to covalently trap azide-modified small molecules in the peptidoglycan cell wall of live mycobacteria after they have traversed the mycomembrane. Using PAC-MAN, we now show that the mycomembrane differentially restricts access to fluorophores and antibiotic derivatives. Mycomembranes of both M. tuberculosis and the model organism Mycobacterium smegmatis discriminate between divergent classes of antibiotics as well as between antibiotics within a single family, the fluoroquinolones. By analyzing subpopulations of M. tuberculosis and M. smegmatis, we also found that some fluorophores and vancomycin are heterogeneously restricted by the mycomembrane. Our data indicate that the mycomembrane is a molecule- and cell-specific barrier to antibiotic permeation.
Collapse
Affiliation(s)
- Irene Lepori
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Kiserian Jackson
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Zichen Liu
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Mahendra D Chordia
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Mitchell Wong
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Sylvia L Rivera
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Marta Roncetti
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
- University of Siena, Siena 53100, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Joel S Freundlich
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Marcos M Pires
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - M Sloan Siegrist
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
4
|
Patel RR, Vidyasagar, Singh SK, Singh M. Recent advances in inhibitor development and metabolic targeting in tuberculosis therapy. Microb Pathog 2025; 203:107515. [PMID: 40154850 DOI: 10.1016/j.micpath.2025.107515] [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: 12/27/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Despite being a preventable and treatable disease, tuberculosis (TB) remained the second leading infectious cause of death globally in 2022, surpassed only by COVID-19. The death rate from TB is influenced by numerous factors that include antibiotic drug resistance, noncompliance with chemotherapy by patients, concurrent infection with the human immunodeficiency virus, delayed diagnosis, varying effectiveness of the Bacille-Calmette-Guerin vaccine, and other factors. Even with the recent advances in our knowledge of Mycobacterium tuberculosis and the accessibility of advanced genomic tools such as proteomics and microarrays, alongside modern methodologies, the pursuit of next-generation inhibitors targeting distinct or multiple molecular pathways remains essential to combat the increasing antimicrobial resistance. Hence, there is an urgent need to identify and develop new drug targets against TB that have unique mechanisms. Novel therapeutic targets might encompass gene products associated with various aspects of mycobacterial biology, such as transcription, metabolism, cell wall formation, persistence, and pathogenesis. This review focuses on the present state of our knowledge and comprehension regarding various inhibitors targeting key metabolic pathways of M. tuberculosis. The discussion encompasses small molecule, synthetic, peptide, natural product and microbial inhibitors and navigates through promising candidates in different phases of clinical development. Additionally, we explore the crucial enzymes and targets involved in metabolic pathways, highlighting their inhibitors. The metabolic pathways explored include nucleotide synthesis, mycolic acid synthesis, peptidoglycan biosynthesis, and energy metabolism. Furthermore, advancements in genetic approaches like CRISPRi and conditional expression systems are discussed, focusing on their role in elucidating gene essentiality and vulnerability in Mycobacteria.
Collapse
Affiliation(s)
- Ritu Raj Patel
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Vidyasagar
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sudhir Kumar Singh
- Virus Research and Diagnostic Laboratory, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Meenakshi Singh
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
5
|
Varshney A, Jia Z, Howe MD, Keiler KC, Baughn AD. A trans-Translation Inhibitor is Potentiated by Zinc and Kills Mycobacterium tuberculosis and Nontuberculous Mycobacteria. ACS Infect Dis 2025; 11:1140-1152. [PMID: 40202906 PMCID: PMC12071686 DOI: 10.1021/acsinfecdis.4c00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Mycobacterium tuberculosis poses a serious challenge for human health, and new antibiotics with novel targets are needed. Here we demonstrate that an acylaminooxadiazole, MBX-4132, specifically inhibits the trans-translation ribosome rescue pathway to kill M. tuberculosis. Our data demonstrate that MBX-4132 is bactericidal against multiple pathogenic mycobacterial species and kills M. tuberculosis in macrophages. We also show that acylaminooxadiazole activity is antagonized by iron but is potentiated by zinc. Our transcriptomic data reveal dysregulation of multiple metal homeostasis pathways after exposure to MBX-4132. Furthermore, we see differential expression of genes related to zinc sensing and efflux when trans-translation is inhibited. Taken together, these data suggest that there is a link between disturbing intracellular metal levels and acylaminooxadiazole-mediated inhibition of trans-translation. These findings provide an important proof-of-concept that trans-translation is a promising antitubercular drug target.
Collapse
Affiliation(s)
- Akanksha Varshney
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Ziyi Jia
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| | - Michael D. Howe
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| | - Kenneth C. Keiler
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Anthony D. Baughn
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
6
|
Xiao G, Cui Y, Zhou L, Niu C, Wang B, Wang J, Zhou S, Pan M, Chan CK, Xia Y, Xu L, Lu Y, Chen S. Identification of a phenyl ester covalent inhibitor of caseinolytic protease and analysis of the ClpP1P2 inhibition in mycobacteria. MLIFE 2025; 4:155-168. [PMID: 40313980 PMCID: PMC12042115 DOI: 10.1002/mlf2.12169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/28/2024] [Accepted: 12/03/2024] [Indexed: 05/03/2025]
Abstract
The caseinolytic protease complex ClpP1P2 is crucial for protein homeostasis in mycobacteria and stress response and virulence of the pathogens. Its role as a potential drug target for combating tuberculosis (TB) has just begun to be substantiated in drug discovery research. We conducted a biochemical screening targeting the ClpP1P2 using a library of compounds phenotypically active against Mycobacterium tuberculosis (Mtb). The screening identified a phenyl ester compound GDI-5755, inhibiting the growth of Mtb and M. bovis BCG, the model organism of mycobacteria. GDI-5755 covalently modified the active-site serine residue of ClpP1, rendering the peptidase inactive, which was delineated through protein mass spectrometry and kinetic analyses. GDI-5755 exerted antibacterial activity by inhibiting ClpP1P2 in the bacteria, which could be demonstrated through a minimum inhibitory concentration (MIC) shift assay with a clpP1 CRISPRi knockdown (clpP1-KD) mutant GH189. The knockdown also remarkably heightened the mutant's sensitivity to ethionamide and meropenem, but not to many other TB drugs. On the other hand, a comparative proteomic analysis of wild-type cells exposed to GDI-5755 revealed the dysregulated proteome, specifically showing changes in the expression levels of multiple TB drug targets, including EthA, LdtMt2, and PanD. Subsequent evaluation confirmed the synergistic activity of GDI-5755 when combined with the TB drugs to inhibit mycobacterial growth. Our findings indicate that small-molecule inhibitors targeting ClpP1P2, when used alongside existing TB medications, could represent novel therapeutic strategies.
Collapse
Affiliation(s)
- Genhui Xiao
- Global Health Drug Discovery InstituteBeijingChina
| | - Yumeng Cui
- Global Health Drug Discovery InstituteBeijingChina
| | | | - Chuya Niu
- Global Health Drug Discovery InstituteBeijingChina
| | - Bing Wang
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, and Beijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Jinglan Wang
- Global Health Drug Discovery InstituteBeijingChina
| | | | - Miaomiao Pan
- Global Health Drug Discovery InstituteBeijingChina
| | - Chi Kin Chan
- Global Health Drug Discovery InstituteBeijingChina
| | - Yan Xia
- Global Health Drug Discovery InstituteBeijingChina
| | - Lan Xu
- Global Health Drug Discovery InstituteBeijingChina
| | - Yu Lu
- Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, and Beijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Shawn Chen
- Global Health Drug Discovery InstituteBeijingChina
| |
Collapse
|
7
|
Fang C, Zhang H, He J, Tian X, Zeng S, Han X, Wang S, Yusuf B, Hu J, Zhong N, Gao Y, Hameed HMA, Zhang T. GrcC1 mediates low-level resistance to multiple drugs in M. marinum, M. abscessus, and M. smegmatis. Microbiol Spectr 2025; 13:e0228924. [PMID: 40009796 PMCID: PMC11960048 DOI: 10.1128/spectrum.02289-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/22/2025] [Indexed: 02/28/2025] Open
Abstract
The escalating threat of mycobacterial infectious diseases, particularly those caused by nontuberculous mycobacteria (NTM), poses a serious challenge to public health. Linezolid (LZD), an oxazolidinone antimicrobial, exhibits potent activity against Mycobacterium tuberculosis and NTM. Generally, mutations in the rrl and rplC genes are widely associated with resistance to LZD. However, in this study, we screened Mycobacterium marinum strains lacking such mutations, indicating the presence of an alternative resistance mechanism. Notably, through whole-genome sequencing, we identified a novel mutation C395T in the MMAR_0911 (grcC1) gene that has never been linked to drug resistance. This mutation leads to an A132V substitution in the encoded protein, a polyprenyl diphosphate synthase potentially involved in the synthesis of cell wall components and menaquinones. We found that the overexpression of grcC1 caused resistance to multiple drugs including LZD, clarithromycin (CLR), vancomycin (VAN), clofazimine (CFZ), rifampicin (RIF), cefoxitin (CEF), levofloxacin (LEV), and moxifloxacin (MXF) and reduced cell wall permeability, while the silence and knockout of grcC1 showed increased cell wall permeability and susceptibility to these drugs. Using CRISPR/Cpf1-assisted gene editing, we confirmed that the A132V mutation conferred low-level resistance to the aforementioned drugs in Mycobacterium abscessus and Mycobacterium smegmatis. Furthermore, thin-layer chromatography analysis indicated reduced glycolipid polarity in the grcC1 mutant strains, suggesting an impact on the cell envelope integrity. Our findings suggest that GrcC1 contributes to low-level drug resistance in mycobacteria by potentially reducing cell wall permeability, highlighting its potential as a novel target for antimicrobial agents and as a diagnostic marker.IMPORTANCEOur study uncovers a novel drug resistance mechanism in mycobacteria, focusing on the previously uncharacterized grcC1 gene. We identified a new mutation, A132V, in GrcC1, which is involved in cell wall component synthesis and menaquinone production. This mutation contributes to low-level resistance not only to linezolid but also to a broad range of drugs, including clarithromycin, vancomycin, and rifampicin. Through advanced techniques like CRISPR interference and gene editing, we demonstrated that GrcC1 plays a critical role in drug susceptibility and cell wall permeability across multiple Mycobacterium species. These findings represent the first connection between GrcC1 and drug resistance, offering new insights into combating infections caused by nontuberculous mycobacteria (NTM). Our work highlights the potential of GrcC1 as a target for novel therapeutic approaches and as a diagnostic marker for drug-resistant NTM infections.
Collapse
Affiliation(s)
- Cuiting Fang
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Han Zhang
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Jing He
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Xirong Tian
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Sanshan Zeng
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Xingli Han
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Shuai Wang
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Buhari Yusuf
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Nanshan Zhong
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Yamin Gao
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - H. M. Adnan Hameed
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- Guangdong-HongKong-Macau Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Oluoch PO, Koh EI, Proulx MK, Reames CJ, Papavinasasundaram KG, Murphy KC, Zimmerman MD, Dartois V, Sassetti CM. Chemical genetic interactions elucidate pathways controlling tuberculosis antibiotic efficacy during infection. Proc Natl Acad Sci U S A 2025; 122:e2417525122. [PMID: 39993187 PMCID: PMC11892619 DOI: 10.1073/pnas.2417525122] [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: 09/04/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025] Open
Abstract
Successful tuberculosis therapy requires treatment with an unwieldy multidrug combination for several months. Thus, there is a growing need to identify novel genetic vulnerabilities that can be leveraged to develop new, more effective antitubercular drugs. Consequently, recent efforts to optimize tuberculosis (TB) therapy have exploited Mycobacterium tuberculosis (Mtb) chemical genetics to identify pathways influencing antibiotic efficacy, novel mechanisms of antibiotic action, and new targets for TB drug discovery. However, the influence of the complex host environment on these interactions remains largely unknown, leaving the therapeutic potential of the identified targets unclear. In this study, we leveraged a library of conditional mutants targeting 467 essential Mtb genes to characterize the chemical-genetic interactions (CGIs) with TB drugs directly in the mouse infection model. We found that these in vivo CGIs differ significantly from those identified in vitro. Both drug-specific and drug-agnostic effects were identified, and many were preserved during treatment with a multidrug combination, suggesting numerous strategies for enhancing therapy. This work also elucidated the complex effects of pyrazinamide (PZA), a drug that relies on aspects of the infection environment for efficacy. Specifically, our work supports the importance of coenzyme A synthesis- inhibition during infection, as well as the antagonistic effect of iron limitation on PZA activity. In addition, we found that inhibition of thiamine and purine synthesis increases PZA efficacy, suggesting additional therapeutically exploitable metabolic dependencies. Our findings present a map of the unique in vivo CGIs, characterizing the mechanism of PZA activity in vivo and identifying potential targets for TB drug development.
Collapse
Affiliation(s)
- Peter O. Oluoch
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Eun-Ik Koh
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Megan K. Proulx
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Charlotte J. Reames
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | | | - Kenan C. Murphy
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Matthew D. Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | | |
Collapse
|
9
|
Xie M, Osiecki P, Rodriguez S, Dartois V, Sarathy J. A Physiologically Relevant In Vitro Model of Nonreplicating Persistent Mycobacterium tuberculosis in Caseum. Curr Protoc 2025; 5:e70118. [PMID: 40056090 PMCID: PMC11890023 DOI: 10.1002/cpz1.70118] [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] [Indexed: 05/13/2025]
Abstract
Tuberculosis (TB) remains one of the leading infectious causes of death worldwide. Persistent bacterial populations in specific microenvironments within the host hamper efficient TB chemotherapy. Caseum in the necrotic core of closed granulomas and cavities of pulmonary TB patients can harbor high burdens of drug-tolerant Mycobacterium tuberculosis (MTB) bacilli, making them particularly difficult to sterilize. Here, we describe protocols for the generation of a surrogate matrix using lipid-rich macrophages to mimic the unique composition of caseum in vivo. Importantly, this caseum surrogate induces metabolic and physiological changes within MTB that reproduce the nonreplicating drug-tolerant phenotype of the pathogen in the native caseous environment, making it advantageous over alternative in vitro models of nonreplicating persistent (NRP) MTB. The protocols include culture of THP-1 monocytes, stimulation of lipid droplet accumulation, lysis and denaturation of the foamy macrophages, inoculation and preadaptation of MTB bacilli in the caseum surrogate, and evaluation of drug bactericidal activity against the NRP population. This novel in vitro model is being used to screen for potent bactericidal antimicrobial agents and to identify vulnerable drug targets, among a variety of other applications, thereby reducing our reliance on in vivo models. © 2025 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Caseum surrogate preparation from γ-irradiated M. tuberculosis-induced foamy THP-1 monocyte-derived macrophages (THPMs) Alternate Protocol 1: Caseum surrogate preparation from stearic acid-induced THPMs Basic Protocol 2: Generation of nonreplicating persistent M. tuberculosis and drug susceptibility testing Alternate Protocol 2: Higher-throughput drug susceptibility screening using caseum surrogate.
Collapse
Affiliation(s)
- Min Xie
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew Jersey
| | - Paulina Osiecki
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew Jersey
| | - Suyapa Rodriguez
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew Jersey
| | - Véronique Dartois
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew Jersey
- Department of Medical SciencesHackensack Meridian School of MedicineNutleyNew Jersey
| | - Jansy Sarathy
- Center for Discovery and InnovationHackensack Meridian HealthNutleyNew Jersey
- Department of Medical SciencesHackensack Meridian School of MedicineNutleyNew Jersey
| |
Collapse
|
10
|
Tran JS, Ward RD, Iruegas-López R, Ebersberger I, Peters JM. Chemical genomics informs antibiotic and essential gene function in Acinetobacter baumannii. PLoS Genet 2025; 21:e1011642. [PMID: 40153700 PMCID: PMC11975115 DOI: 10.1371/journal.pgen.1011642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/07/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025] Open
Abstract
The Gram-negative pathogen, Acinetobacter baumannii, poses a serious threat to human health due to its role in nosocomial infections that are resistant to treatment with current antibiotics. Despite this, our understanding of fundamental A. baumannii biology remains limited, as many essential genes have not been experimentally characterized. These essential genes are critical for bacterial survival and, thus, represent promising targets for drug discovery. Here, we systematically probe the function of essential genes by screening a CRISPR interference knockdown library against a diverse panel of chemical inhibitors, including antibiotics. We find that most essential genes show chemical-gene interactions, allowing insights into both inhibitor and gene function. For instance, knockdown of lipooligosaccharide (LOS) transport genes increased sensitivity to a broad range of chemicals. Cells with defective LOS transport showed cell envelope hyper-permeability that was dependent on continued LOS synthesis. Using phenotypes across our chemical-gene interaction dataset, we constructed an essential gene network linking poorly understood genes to well-characterized genes in cell division and other processes. Finally, our phenotype-structure analysis identified structurally related antibiotics with distinct cellular impacts and suggested potential targets for underexplored inhibitors. This study advances our understanding of essential gene and inhibitor function, providing a valuable resource for mechanistic studies, therapeutic strategies, and future key targets for antibiotic development.
Collapse
Affiliation(s)
- Jennifer Suzanne Tran
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ryan David Ward
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rubén Iruegas-López
- Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Ingo Ebersberger
- Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt am Main, Germany
| | - Jason Matthew Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
11
|
Lepori I, Liu Z, Evbarunegbe N, Feng S, Brown TP, Mane K, Shivangi, Wong M, George A, Guo T, Dong J, Freundlich JS, Im W, Green AG, Pires MM, Siegrist MS. Identification of chemical features that influence mycomembrane permeation and antitubercular activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640664. [PMID: 40060676 PMCID: PMC11888444 DOI: 10.1101/2025.02.27.640664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is the deadliest single-agent infection worldwide. Current antibiotic treatment for TB takes a minimum of four months, underscoring the need for better therapeutics. The unique mycobacterial cell envelope, particularly the outermost mycomembrane, has long been thought to promote intrinsic antibiotic resistance by limiting compound entry into Mtb. Understanding chemical features that influence permeation across the mycomembrane may enable more accurate predictions of whole cell anti-Mtb activity, leading to development of more effective antibacterials. Here we query the mycomembrane permeation of over 1500 azide-tagged compounds in live Mtb with the bioorthogonal click chemistry-based assay PAC-MAN. We use cheminformatics and machine learning to identify chemical features associated with mycomembrane permeation and show that they have predictive value via systematic modification of two small molecule series. Additionally, we find that chemical features that influence mycomembrane permeation correlate with anti-Mtb activity in large compound libraries. These findings suggest that the mycomembrane is indeed a significant barrier to whole cell activity in Mtb and provide a rational framework for designing or modifying compounds to overcome this barrier.
Collapse
Affiliation(s)
- Irene Lepori
- Department of Microbiology, University of Massachusetts, Amherst; Amherst, MA, USA
| | - Zichen Liu
- Department of Chemistry, University of Virginia; Charlottesville, VA, USA
| | - Nelson Evbarunegbe
- Manning College of Information and Computer Sciences, University of Massachusetts, Amherst; Amherst, MA, USA
| | - Shasha Feng
- Department of Biological Sciences, Lehigh University; Bethlehem, PA, USA
- Department of Bioengineering, Lehigh University; Bethlehem, PA, USA
| | - Turner P. Brown
- Department of Biological Sciences, Lehigh University; Bethlehem, PA, USA
- Department of Bioengineering, Lehigh University; Bethlehem, PA, USA
| | - Kishor Mane
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School; Newark, NJ, USA
| | - Shivangi
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School; Newark, NJ, USA
| | - Mitchell Wong
- Department of Microbiology, University of Massachusetts, Amherst; Amherst, MA, USA
| | - Amir George
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School; Newark, NJ, USA
| | - Taijie Guo
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University; Shanghai, China
| | - Jiajia Dong
- Institute of Translational Medicine, Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University; Shanghai, China
| | - Joel S. Freundlich
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School; Newark, NJ, USA
| | - Wonpil Im
- Department of Biological Sciences, Lehigh University; Bethlehem, PA, USA
- Department of Bioengineering, Lehigh University; Bethlehem, PA, USA
| | - Anna G. Green
- Manning College of Information and Computer Sciences, University of Massachusetts, Amherst; Amherst, MA, USA
| | - Marcos M. Pires
- Department of Chemistry, University of Virginia; Charlottesville, VA, USA
| | - M. Sloan Siegrist
- Department of Microbiology, University of Massachusetts, Amherst; Amherst, MA, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst; Amherst, MA, USA
| |
Collapse
|
12
|
Dong J, Cheng Q, Tang C, Zhong Y, Wang J, Lv M, Chen Z, Li P, Luo M, Pei H. Comparative In Vitro Drug Susceptibility Study of Five Oxazolidinones Against Mycobacterium tuberculosis in Hainan, China. Pathogens 2025; 14:218. [PMID: 40137704 PMCID: PMC11945096 DOI: 10.3390/pathogens14030218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Oxazolidinones, novel synthetic antibacterials, inhibit protein biosynthesis and show potent activity against Gram-positive bacteria, including Mycobacterium tuberculosis (MTB). In this study, we aimed to compare the in vitro activity of linezolid (LZD) and four oxazolidinones, including tedizolid (TZD), contezolid (CZD), sutezolid (SZD), and delpazolid (DZD), against multidrug-resistant tuberculosis (MDR-TB) and pre-extensively drug-resistant tuberculosis (pre-XDR-TB) isolates from Hainan. We established their epidemiological cut-off values (ECOFFs) using ECOFFinder software and analyzed mutations in rrl (23S rRNA), rplC, rplD, mce3R, tsnR, Rv0545c, Rv0930, Rv3331, and Rv0890c genes to uncover potential mechanisms of oxazolidinone resistance. This study included 177 MTB isolates, comprising 67 MDR and 110 pre-XDR-TB isolates. Overall, SZD exhibited the strongest antibacterial activity against clinical MTB isolates, followed by TZD and LZD, with CZD and DZD showing equivalent but weaker activity (SZDMIC50 = TZDMIC50 < LZDMIC50 < CZDMIC50 = DZDMIC50; SZDMIC90 < TZDMIC90 = LZDMIC90 < CZDMIC90 = DZDMIC90). Significant differences in MIC distribution were observed for TZD (p < 0.0001), CZD (p < 0.01), SZD (p < 0.0001), and DZD (p < 0.0001) compared to LZD but not between MDR-TB and pre-XDR-TB isolates. We propose the following ECOFFs: SZD, 0.5 µg/mL; LZD, TZD, and CZD, 1.0 µg/mL; DZD, 2.0 µg/mL. No statistically significant differences in resistance rates were observed among these five drugs (p > 0.05). We found that eight MTB isolates (4.52% [8/177]) resisted these five oxazolidinones. Among these, only one isolate, M26, showed an amino acid substitution (Arg79His) in the protein encoded by the rplD gene, which conferred cross-resistance to TZD and CZD. Three distinct mutations were identified in the mce3R gene; notably, isolate P604 displayed two insertions that contributed to resistance against all five oxazolidinones. However, no significant correlation was observed between mutations in the rrl, rplC, rplD, mce3R, tsnR, Rv0545c, Rv0930, Rv3331, and Rv0890c genes with oxazolidinone resistance in the clinical MTB isolates tested. In summary, this study provides the first report on the resistance of MTB in Hainan to the five oxazolidinones (LZD, TZD, CZD, SZD, and DZD). In vitro susceptibility testing indicated that SZD exhibited the strongest antibacterial activity, followed by TZD and LZD, while CZD and DZD demonstrated comparable but weaker effectiveness. Mutations in rplD and mce3R were discovered, but further research is needed to clarify their role in conferring oxazolidinone resistance in MTB.
Collapse
Affiliation(s)
- Jinhui Dong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| | - Qian Cheng
- Tuberculosis Research Units, Chongqing Public Health Medical Center, Chongqing 400036, China; (Q.C.); (P.L.)
| | - Chuanning Tang
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou 571199, China;
| | - Yeteng Zhong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| | - Jieying Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| | - Meiping Lv
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| | - Zhuolin Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| | - Peibo Li
- Tuberculosis Research Units, Chongqing Public Health Medical Center, Chongqing 400036, China; (Q.C.); (P.L.)
| | - Ming Luo
- Clinical Laboratory, Chongqing Public Health Medical Center, Chongqing 400036, China
| | - Hua Pei
- Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou 571199, China; (J.D.); (Y.Z.); (J.W.); (M.L.); (Z.C.)
| |
Collapse
|
13
|
Laudouze J, Point V, Achache W, Crauste C, Canaan S, Santucci P. Fluorescence-based CRISPR interference system for controlled genetic repression and live single-cell imaging in mycobacteria. FEBS Lett 2025; 599:488-501. [PMID: 39618159 PMCID: PMC11848015 DOI: 10.1002/1873-3468.15071] [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: 10/06/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 02/25/2025]
Abstract
In this research letter, we report the development and validation of a new subset of fluorescence-based CRISPR interference (CRISPRi) tools for our scientific community. The pJL series is directly derived from the original pIRL CRISPRi vectors and conserves all the elements to perform inducible targeted gene repression. These vectors carry two distinct fluorescent markers under the constitutive promoter psmyc to simplify the selection of recombinant clones. We demonstrate the functionality of these vectors by targeting the expression of the glycopeptidolipid translocase mmpL4b and the essential genes rpoB and mmpL3. Finally, we describe an efficient single-step procedure to co-transform mycobacterial species with this integrative genetic tool alongside episomal vectors. Such tools and approaches should be useful to foster discovery in mycobacterial research.
Collapse
Affiliation(s)
| | - Vanessa Point
- Aix Marseille Univ, CNRS, LISM, IMM FR3479, IM2BFrance
| | - Wafaa Achache
- Aix Marseille Univ, CNRS, LISM, IMM FR3479, IM2BFrance
- IHU Méditerranée Infection, Aix‐Marseille Univ.France
| | | | | | | |
Collapse
|
14
|
Zhang J, Ju Y, Li L, Hameed HMA, Yusuf B, Gao Y, Fang C, Tian X, Ding J, Ma W, Chen X, Wang S, Zhang T. MtrAB two-component system is crucial for the intrinsic resistance and virulence of Mycobacterium abscessus. Int J Antimicrob Agents 2025; 65:107442. [PMID: 39761758 DOI: 10.1016/j.ijantimicag.2024.107442] [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: 04/17/2024] [Revised: 12/04/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025]
Abstract
Mycobacterium abscessus (Mab) poses serious therapeutic challenges, largely due to its intrinsic resistance to many antibiotics. The development of targeted therapeutic strategies necessitates the identification of bacterial factors that contribute to its reduced susceptibility to antibiotics and/or to the killing by its host cells. In this study, we discovered that Mab strains with disrupted mtrA, mtrB or both, or a gene-edited mtrA encoding MtrA with Tyr102Cys mutation, exhibited highly increased sensitivity to various drugs compared to the wild-type Mab. In a murine model, three antibiotics inactive against the wild-type Mab demonstrated efficacy against the mtrA and mtrB knockout strains, significantly reducing pulmonary bacterial burdens compared to untreated controls. Notably, the virulence of all the mtrA, mtrB and mtrAB knockout mutants was highly diminished, evidenced by a reduced bacterial load in mouse lungs, undetectable level in spleens, and defective growth in macrophage RAW264.7. Morphological analysis revealed elongated cell length and multiple septa in knockout strains, suggesting both MtrA and MtrB regulate cell division of Mab. Furthermore, the absence of mtrA, mtrB or both significantly increased cell envelope permeability and reduced biofilm formation. Transcriptome sequencing showed altered expression levels of multiple genes related to plasma membrane, fatty acid metabolism and biosynthesis pathways in wild-type Mab and mtrA knockout strain. In summary, this study suggests that MtrA and MtrB play a crucial role in the intrinsic resistance and virulence of Mab by affecting cell division and altering cell permeability. Consequently, MtrA and MtrB represent promising targets for the discovery of anti-Mab drugs.
Collapse
Affiliation(s)
- Jingran Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - Yanan Ju
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - Lijie Li
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - H M Adnan Hameed
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - Buhari Yusuf
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; University of Chinese Academy of Science, Beijing, China
| | - Yamin Gao
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - Cuiting Fang
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; University of Chinese Academy of Science, Beijing, China
| | - Xirong Tian
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; University of Chinese Academy of Science, Beijing, China
| | - Jie Ding
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Wanli Ma
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; University of Chinese Academy of Science, Beijing, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, China
| | - Shuai Wang
- Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China.
| | - Tianyu Zhang
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Institute of Drug Discovery, State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China; University of Chinese Academy of Science, Beijing, China; Guangzhou National Laboratory, Guangzhou, China.
| |
Collapse
|
15
|
Sweeney MI, Carranza CE, Tobin DM. Understanding Mycobacterium tuberculosis through its genomic diversity and evolution. PLoS Pathog 2025; 21:e1012956. [PMID: 40019877 PMCID: PMC11870338 DOI: 10.1371/journal.ppat.1012956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025] Open
Abstract
Pathogen evolution and genomic diversity are shaped by specific host immune pressures and therapeutic interventions. Analysis of the extant genomes of circulating strains of Mycobacterium tuberculosis, a leading cause of infectious mortality that has co-evolved with humans for thousands of years, can provide new insights into host-pathogen interactions that underlie specific aspects of pathogenesis and onward transmission. With the explosion in the number of fully sequenced M. tuberculosis strains that are now paired with detailed clinical data, there are new opportunities to understand the evolutionary basis for and consequences of M. tuberculosis strain diversity. This review examines mechanistic findings that have emerged from pairing whole genome sequencing data and evolutionary analysis with functional dissection of specific bacterial variants. These include improved understanding of secreted effectors that modulate the properties and migratory behavior of infected macrophages as well as bacterial genetic alterations important for survival within hypoxic microenvironments. Genomic, evolutionary, and functional analyses across diverse M. tuberculosis strains will identify prominent bacterial adaptations to their human hosts and shape our understanding of TB disease biology and the host immune response.
Collapse
Affiliation(s)
- Mollie I. Sweeney
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Carson E. Carranza
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
16
|
Bagchi S, Sharma AK, Mal S, Kundu M, Basu J. Crosstalk between cyclic-di-guanosine monophosphate and the sensor kinase MtrB regulates MtrA-dependent genes, bacterial growth, biofilm formation and lysosomal trafficking of Mycobacterium tuberculosis. MICROBIOLOGY (READING, ENGLAND) 2025; 171. [PMID: 39918987 DOI: 10.1099/mic.0.001532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Cyclic-di-guanosine monophosphate (c-di-GMP) plays an important role in bacterial signalling networks. C-di-GMP exerts a regulatory function through binding to diverse molecules that include transcription factors, riboswitches and sensor kinases (SKs), thereby regulating diverse processes. Here, we demonstrate the crosstalk between c-di-GMP and the SK MtrB of Mycobacterium tuberculosis. MtrB phosphorylates and regulates its cognate response regulator MtrA. C-di-GMP binds directly to the cytosolic domain of MtrB to inhibit its autophosphorylation. C-di-GMP levels in M. tuberculosis were manipulated by overexpressing a c-di-GMP synthesizing enzyme ydeH and a degrading enzyme rv1357c. We demonstrate that overexpression of ydeH lowers growth of the bacterium both in vitro and in M. tuberculosis grown in macrophages. This is in conformity with lowered expression of mtrA and selected genes of the mtrA regulon involved in cell wall turnover in the ydeH-overexpressing strain compared to the parent strain. We also demonstrate that overexpression of ydeH in M. tuberculosis hinders biofilm formation, whereas overexpression of rv1357c has the opposite effect. Neither of the two genes could rescue the biofilm defective phenotype of the MtrB knock out mutant (ΔmtrB), suggesting that c-di-GMP exerts its role on biofilm formation through MtrB. Finally, we show by fluorescence microscopy that the trafficking of M. tuberculosis overexpressing ydeH is significantly higher than that of the parent strain and that this is linked to reduced expression of the MtrB-dependent genes esxG and esxH, which play a role in subversion of lysosomal trafficking of M. tuberculosis. These results provide important new insight into the crosstalk between c-di-GMP and MtrB in M. tuberculosis.
Collapse
Affiliation(s)
- Shreya Bagchi
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Arun Kumar Sharma
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Soumya Mal
- Department of Biological Sciences, Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, India
| | - Manikuntala Kundu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| | - Joyoti Basu
- Department of Chemical Sciences, Bose Institute, 93/1 APC Road, Kolkata 700009, India
| |
Collapse
|
17
|
Lepori I, Jackson K, Liu Z, Chordia MD, Wong M, Rivera SL, Roncetti M, Poliseno L, Pires MM, Siegrist MS. The mycomembrane differentially and heterogeneously restricts antibiotic permeation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630956. [PMID: 39803531 PMCID: PMC11722283 DOI: 10.1101/2024.12.31.630956] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The recalcitrance of Mycobacterium tuberculosis to antibiotic treatment has been broadly attributed to the impermeability of the organism's outer mycomembrane. However, the studies that support this inference have been indirect and/or reliant on bulk population measurements. We previously developed the P eptidoglycan A ccessibility C lick- M ediated A ssessme N t (PAC-MAN) method to covalently trap azide-modified small molecules in the peptidoglycan cell wall of live mycobacteria, after they have traversed the mycomembrane. Using PAC-MAN we now show that the mycomembrane differentially restricts access of fluorophores and antibiotic derivatives. Mycomembranes of both M. tuberculosis and the model organism M. smegmatis discriminate between divergent classes of antibiotics as well as between antibiotics within a single family, the fluoroquinolones. By analyzing sub-populations of M. tuberculosis and M. smegmatis , we also found that some fluorophores and vancomycin are heterogeneously restricted by the mycomembrane. Our data indicate that the mycomembrane is a molecule- and cell-specific barrier to antibiotic permeation. Abstract Graphic
Collapse
|
18
|
Dartois V, Dick T. Toward better cures for Mycobacterium abscessus lung disease. Clin Microbiol Rev 2024; 37:e0008023. [PMID: 39360834 PMCID: PMC11629636 DOI: 10.1128/cmr.00080-23] [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] [Indexed: 10/30/2024] Open
Abstract
SUMMARYThe opportunistic pathogen Mycobacterium abscessus (Mab) causes fatal lung infections that bear similarities-and notable differences-with tuberculosis (TB) pulmonary disease. In contrast to TB, no antibiotic is formally approved to treat Mab disease, there is no reliable cure, and the discovery and development pipeline is incredibly thin. Here, we discuss the factors behind the unsatisfactory cure rates of Mab disease, namely intrinsic resistance and persistence of the pathogen, and the use of underperforming, often parenteral and toxic, repurposed drugs. We propose preclinical strategies to build injectable-free sterilizing and safe regimens: (i) prioritize oral bactericidal antibiotic classes, with an initial focus on approved agents or advanced clinical candidates to provide immediate options for desperate patients, (ii) test drug combinations early, (iii) optimize novel leads specifically for M. abscessus, and (iv) consider pharmacokinetic-pharmacodynamic targets at the site of disease, the lung lesions in which drug tolerant bacterial populations reside. Knowledge and tool gaps in the preclinical drug discovery process are identified, including validated mouse models and computational platforms to enable in vitro mouse-human translation. We briefly discuss recent advances in clinical development, the need for readouts and biomarkers that correlate with cure, and clinical trial concepts adapted to the uniqueness of Mab patient populations for new regimen development. In an era when most pharmaceutical firms have withdrawn from antimicrobial drug discovery, the breakthroughs needed to fill the regimen development pipeline will likely come from partnerships between academia, biotech, pharma, non-profit organizations, and governments, with incentives that reward cooperation.
Collapse
Affiliation(s)
- Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
19
|
Tran JS, Ward RD, Iruegas-López R, Ebersberger I, Peters JM. Chemical genomics informs antibiotic and essential gene function in Acinetobacter baumannii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627103. [PMID: 39677645 PMCID: PMC11643038 DOI: 10.1101/2024.12.05.627103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The Gram-negative pathogen, Acinetobacter baumannii , poses a serious threat to human health due to its role in nosocomial infections that are resistant to treatment with current antibiotics. Despite this, our understanding of fundamental A. baumannii biology remains limited, as many essential genes have not been experimentally characterized. These essential genes are critical for bacterial survival and, thus, represent promising targets for drug discovery. Here, we systematically probe the function of essential genes by screening a CRISPR interference knockdown library against a diverse panel of chemical inhibitors, including antibiotics. We find that most essential genes show chemical-gene interactions, allowing insights into both inhibitor and gene function. For instance, knockdown of lipooligosaccharide (LOS) transport genes increased sensitivity to a broad range of chemicals. Cells with defective LOS transport showed cell envelope hyper-permeability that was dependent on continued LOS synthesis. Using phenotypes across our chemical-gene interaction dataset, we constructed an essential gene network linking poorly understood genes to well-characterized genes in cell division and other processes. Finally, our phenotype-structure analysis identified structurally related antibiotics with distinct cellular impacts and suggested potential targets for underexplored inhibitors. This study advances our understanding of essential gene and inhibitor function, providing a valuable resource for mechanistic studies, therapeutic strategies, and future key targets for antibiotic development.
Collapse
|
20
|
Rahman ASMZ, Syroegin EA, Novomisky Nechcoff J, Devarajan A, Polikanov YS, Cardona ST. Rationally designed pooled CRISPRi-seq uncovers an inhibitor of bacterial peptidyl-tRNA hydrolase. Cell Rep 2024; 43:114967. [PMID: 39541213 DOI: 10.1016/j.celrep.2024.114967] [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: 06/28/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Bacterial mutant libraries with downregulated antibiotic targets are useful tools for elucidating the mechanisms of action of antibacterial compounds, a pivotal step in antibiotic discovery. However, achieving genomic coverage of antibacterial targets poses a challenge due to the uneven proliferation of knockdown mutants during pooled growth, leading to the unintended loss of important targets. To overcome this issue, we constructed an arrayed essential gene mutant library (EGML) in the antibiotic-resistant bacterium Burkholderia cenocepacia using CRISPR interference (CRISPRi). By modeling depletion levels and adjusting knockdown mutant inocula, we rationally designed and optimized a CRISPR interference-mediated pooled library of essential genes (CIMPLE) approaching coverage of the bacterial essential genome with mutant sensitization. We exposed CIMPLE to an uncharacterized bacterial growth inhibitor structurally different from antibiotics and discovered that it inhibits the essential peptidyl-tRNA hydrolase. Overall, CIMPLE leverages the advantages of arrayed and pooled CRISPRi libraries to uncover unexplored targets for antibiotic action.
Collapse
Affiliation(s)
- A S M Zisanur Rahman
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Egor A Syroegin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | - Archit Devarajan
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA; Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA; Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Silvia T Cardona
- Department of Microbiology, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
21
|
Ofori-Anyinam B, Hamblin M, Coldren ML, Li B, Mereddy G, Shaikh M, Shah A, Grady C, Ranu N, Lu S, Blainey PC, Ma S, Collins JJ, Yang JH. Catalase activity deficiency sensitizes multidrug-resistant Mycobacterium tuberculosis to the ATP synthase inhibitor bedaquiline. Nat Commun 2024; 15:9792. [PMID: 39537610 PMCID: PMC11561320 DOI: 10.1038/s41467-024-53933-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Multidrug-resistant tuberculosis (MDR-TB), defined as resistance to the first-line drugs isoniazid and rifampin, is a growing source of global mortality and threatens global control of tuberculosis disease. The diarylquinoline bedaquiline has recently emerged as a highly efficacious drug against MDR-TB and kills Mycobacterium tuberculosis by inhibiting mycobacterial ATP synthase. However, the mechanisms underlying bedaquiline's efficacy against MDR-TB remain unknown. Here we investigate bedaquiline hyper-susceptibility in drug-resistant Mycobacterium tuberculosis using systems biology approaches. We discovered that MDR clinical isolates are commonly sensitized to bedaquiline. This hypersensitization is caused by several physiological changes induced by deficient catalase activity. These include enhanced accumulation of reactive oxygen species, increased susceptibility to DNA damage, induction of sensitizing transcriptional programs, and metabolic repression of several biosynthetic pathways. In this work we demonstrate how resistance-associated changes in bacterial physiology can mechanistically induce collateral antimicrobial drug sensitivity and reveal druggable vulnerabilities in antimicrobial resistant pathogens.
Collapse
Affiliation(s)
- Boatema Ofori-Anyinam
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Meagan Hamblin
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Eversana Consulting, Boston, MA, 02120, USA
| | - Miranda L Coldren
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98105, USA
| | - Barry Li
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Gautam Mereddy
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Mustafa Shaikh
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Avi Shah
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Courtney Grady
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Public Health Research Institute, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Navpreet Ranu
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- insitro, South San Francisco, CA, 94080, USA
| | - Sean Lu
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Paul C Blainey
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Koch Institute of Integrative Cancer Research at MIT, Cambridge, MA, 02139, USA
| | - Shuyi Ma
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98105, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
- Pathobiology Graduate Program, Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - James J Collins
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jason H Yang
- Ruy V. Lourenço Center for Emerging and Re-Emerging Pathogens, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
22
|
Islam T, Josephs EA. Genome editing outcomes reveal mycobacterial NucS participates in a short-patch repair of DNA mismatches. Nucleic Acids Res 2024; 52:12295-12307. [PMID: 38747340 PMCID: PMC11551744 DOI: 10.1093/nar/gkae402] [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: 11/13/2023] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024] Open
Abstract
In the canonical DNA mismatch repair (MMR) mechanism in bacteria, if a nucleotide is incorrectly mis-paired with the template strand during replication, the resulting repair of this mis-pair can result in the degradation and re-synthesis of hundreds or thousands of nucleotides on the newly-replicated strand (long-patch repair). While mycobacteria, which include important pathogens such as Mycobacterium tuberculosis, lack the otherwise highly-conserved enzymes required for the canonical MMR reaction, it was found that disruption of a mycobacterial mismatch-sensitive endonuclease NucS results in a hyper-mutative phenotype, leading to the idea that NucS might be involved in a cryptic, independently-evolved DNA MMR mechanism, perhaps mediated by homologous recombination (HR) with a sister chromatid. Using oligonucleotide recombination, which allows us to introduce mismatches specifically into the genomes of a model for M. tuberculosis, Mycobacterium smegmatis, we find that NucS participates in a direct repair of DNA mismatches where the patch of excised nucleotides is largely confined to within ∼5-6 bp of the mis-paired nucleotides, which is inconsistent with mechanistic models of canonical mycobacterial HR or other double-strand break (DSB) repair reactions. The results presented provide evidence of a novel NucS-associated mycobacterial MMR mechanism occurring in vivo to regulate genetic mutations in mycobacteria.
Collapse
Affiliation(s)
- Tanjina Islam
- Department of Nanoscience, University of North Carolina at Greensboro, Greensboro, NC 27401, USA
| | - Eric A Josephs
- Department of Nanoscience, University of North Carolina at Greensboro, Greensboro, NC 27401, USA
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC 27401, USA
| |
Collapse
|
23
|
Walker AS, Clardy J. Primed for Discovery. Biochemistry 2024; 63:2705-2713. [PMID: 39497571 PMCID: PMC11542185 DOI: 10.1021/acs.biochem.4c00464] [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: 08/10/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/09/2024]
Abstract
Antibiotics are essential components of current medical practice, but their effectiveness is being eroded by the increasing emergence of antimicrobial-resistant infections. At the same time, the rate of antibiotic discovery has slowed, and our future ability to treat infections is threatened. Among Christopher T. Walsh's many contributions to science was his early recognition of this threat and the potential of biosynthesis─genes and mechanisms─to contribute solutions. Here, we revisit a 2006 review by Walsh and co-workers that highlighted a major challenge in antibiotic natural product discovery: the daunting odds for identifying new naturally occurring antibiotics. The review described strategies to mitigate the odds challenge. These strategies have been used extensively by the natural product discovery community in the years since and have resulted in some promising discoveries. Despite these advances, the rarity of novel antibiotic natural products remains a barrier to discovery. We compare the challenge of discovering natural product antibiotics to the process of identifying new prime numbers, which are also challenging to find and an essential, if underappreciated, element of modern life. We propose that inclusion of filters for functional compounds early in the discovery pipeline is key to natural product antibiotic discovery, review some recent advances that enable this, and discuss some remaining challenges that need to be addressed to make antibiotic discovery sustainable in the future.
Collapse
Affiliation(s)
- Allison S. Walker
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
24
|
Romano KP, Bagnall J, Warrier T, Sullivan J, Ferrara K, Orzechowski M, Nguyen PH, Raines K, Livny J, Shoresh N, Hung DT. Perturbation-specific transcriptional mapping for unbiased target elucidation of antibiotics. Proc Natl Acad Sci U S A 2024; 121:e2409747121. [PMID: 39467118 PMCID: PMC11551328 DOI: 10.1073/pnas.2409747121] [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: 06/10/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
The rising prevalence of antibiotic resistance threatens human health. While more sophisticated strategies for antibiotic discovery are being developed, target elucidation of new chemical entities remains challenging. In the postgenomic era, expression profiling can play an important role in mechanism-of-action (MOA) prediction by reporting on the cellular response to perturbation. However, the broad application of transcriptomics has yet to fulfill its promise of transforming target elucidation due to challenges in identifying the most relevant, direct responses to target inhibition. We developed an unbiased strategy for MOA prediction, called perturbation-specific transcriptional mapping (PerSpecTM), in which large-throughput expression profiling of wild-type or hypomorphic mutants, depleted for essential targets, enables a computational strategy to address this challenge. We applied PerSpecTM to perform reference-based MOA prediction based on the principle that similar perturbations, whether chemical or genetic, will elicit similar transcriptional responses. Using this approach, we elucidated the MOAs of three molecules with activity against Pseudomonas aeruginosa by comparing their expression profiles to those of a reference set of antimicrobial compounds with known MOAs. We also show that transcriptional responses to small-molecule inhibition resemble those resulting from genetic depletion of essential targets by clustered regularly interspaced short palindromic repeats interference (CRISPRi) by PerSpecTM, demonstrating proof of concept that correlations between expression profiles of small-molecule and genetic perturbations can facilitate MOA prediction when no chemical entities exist to serve as a reference. Empowered by PerSpecTM, this work lays the foundation for an unbiased, readily scalable, systematic reference-based strategy for MOA elucidation that could transform antibiotic discovery efforts.
Collapse
Affiliation(s)
- Keith P. Romano
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Josephine Bagnall
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Thulasi Warrier
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
| | - Jaryd Sullivan
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Genetics, Harvard Medical School, Boston, MA02115
| | - Kristina Ferrara
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
| | - Marek Orzechowski
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Phuong H. Nguyen
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Genetics, Harvard Medical School, Boston, MA02115
| | - Kyra Raines
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
| | - Jonathan Livny
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Noam Shoresh
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Deborah T. Hung
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Genetics, Harvard Medical School, Boston, MA02115
| |
Collapse
|
25
|
De Boeck N, Villellas C, Crespo-Yuste E, Gonzalo-Asensio J, Buckley PT, Thys K, Vuong C, Lounis N, Verstraeten N, Michiels J. A single upstream mutation of whiB7 underlies amikacin and clarithromycin resistance in Mycobacterium abscessus. J Appl Microbiol 2024; 135:lxae286. [PMID: 39537195 DOI: 10.1093/jambio/lxae286] [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: 08/08/2024] [Revised: 10/18/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024]
Abstract
AIMS We aimed to investigate the molecular mechanisms underlying the survival of Mycobacterium abscessus when faced with antibiotic combination therapy. By conducting evolution experiments and whole-genome sequencing (WGS), we sought to identify genetic variants associated with stress response mechanisms, with a particular focus on drug survival and resistance. METHODS AND RESULTS We conducted evolution experiments on M. abscessus, exposing the bacteria to a combination therapy of amikacin and rifabutin. Genetic mutations associated with increased antibiotic survival and altered susceptibility were subsequently identified by WGS. We focused on mutations that contribute to stress response mechanisms and tolerance. Of particular interest was a novel frameshift mutation in MAB_3509c, a gene of unknown function within the upstream open reading frame of whiB7. A MAB_3509c knockout mutant was constructed, and expression of downstream drug resistance genes was assessed by RT-qPCR. Mutation of MAB_3509c results in increased RNA levels of whiB7 and downstream stress response genes such as eis2, which is responsible for aminoglycoside resistance. CONCLUSION Our findings demonstrate the importance of whiB7 in the adaptive stress response in M. abscessus. Moreover, our results highlight the complexity of M. abscessus adapting to drug stress and underscore the need for further research.
Collapse
Affiliation(s)
- Nathan De Boeck
- Center for Microbiology, VIB-KU Leuven, 3001 Leuven, Belgium
- Centre of Microbial and Plant Genetics, KU Leuven, 3001 Leuven, Belgium
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Cristina Villellas
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Facultad de Medicina, Universidad de Zaragoza IIS-Aragón, 50009 Zaragoza, Spain
| | - Estefanía Crespo-Yuste
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Facultad de Medicina, Universidad de Zaragoza IIS-Aragón, 50009 Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesús Gonzalo-Asensio
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Facultad de Medicina, Universidad de Zaragoza IIS-Aragón, 50009 Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Peter T Buckley
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Kim Thys
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Cuong Vuong
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Nacer Lounis
- Infectious Diseases Therapeutic Area, LLC, Janssen Pharmaceutica NV, 2340 Beerse, Belgium
| | - Natalie Verstraeten
- Center for Microbiology, VIB-KU Leuven, 3001 Leuven, Belgium
- Centre of Microbial and Plant Genetics, KU Leuven, 3001 Leuven, Belgium
| | - Jan Michiels
- Center for Microbiology, VIB-KU Leuven, 3001 Leuven, Belgium
- Centre of Microbial and Plant Genetics, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
26
|
Varshney A, Jia Z, Howe MD, Keiler KC, Baughn AD. A trans-translation inhibitor is potentiated by zinc and kills Mycobacterium tuberculosis and non-tuberculous mycobacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.02.621434. [PMID: 39554143 PMCID: PMC11566007 DOI: 10.1101/2024.11.02.621434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Mycobacterium tuberculosis poses a serious challenge for human health, and new antibiotics with novel targets are needed. Here we demonstrate that an acylaminooxadiazole, MBX-4132, specifically inhibits the trans-translation ribosome rescue pathway to kill M. tuberculosis. Our data demonstrate that MBX-4132 is bactericidal against multiple pathogenic mycobacterial species and kills M. tuberculosis in macrophages. We also show that acylaminooxadiazole activity is antagonized by iron but is potentiated by zinc. Our transcriptomic data reveals dysregulation of multiple metal homeostasis pathways after exposure to MBX-4132. Furthermore, we see differential expression of genes related to zinc sensing and efflux when trans-translation is inhibited. Taken together, these data suggest that there is a link between disturbing intracellular metal levels and acylaminooxadiazole-mediated inhibition of trans-translation. These findings provide an important proof-of-concept that trans-translation is a promising antitubercular drug target.
Collapse
Affiliation(s)
- Akanksha Varshney
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
- These authors contributed equally
| | - Ziyi Jia
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455 USA
- These authors contributed equally
| | - Michael D. Howe
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455 USA
| | - Kenneth C. Keiler
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712 USA
| | - Anthony D. Baughn
- Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455 USA
| |
Collapse
|
27
|
Kim LM, Todor H, Gross CA. Correction of a widespread bias in pooled chemical genomics screens improves their interpretability. Mol Syst Biol 2024; 20:1173-1186. [PMID: 39349762 PMCID: PMC11535069 DOI: 10.1038/s44320-024-00069-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: 05/30/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 11/06/2024] Open
Abstract
Chemical genomics is a powerful and increasingly accessible technique to probe gene function, gene-gene interactions, and antibiotic synergies and antagonisms. Indeed, multiple large-scale pooled datasets in diverse organisms have been published. Here, we identify an artifact arising from uncorrected differences in the number of cell doublings between experiments within such datasets. We demonstrate that this artifact is widespread, show how it causes spurious gene-gene and drug-drug correlations, and present a simple but effective post hoc method for removing its effects. Using several published datasets, we demonstrate that this correction removes spurious correlations between genes and conditions, improving data interpretability and revealing new biological insights. Finally, we determine experimental factors that predispose a dataset for this artifact and suggest a set of experimental and computational guidelines for performing pooled chemical genomics experiments that will maximize the potential of this powerful technique.
Collapse
Affiliation(s)
- Lili M Kim
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Horia Todor
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Carol A Gross
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
- California Institute of Quantitative Biology, University of California, San Francisco, San Francisco, 94158, CA, USA
| |
Collapse
|
28
|
Singpanomchai N, Ratthawongjirakul P. The CRISPR-dCas9 interference system suppresses inhA gene expression in Mycobacterium smegmatis. Sci Rep 2024; 14:26116. [PMID: 39478003 PMCID: PMC11525817 DOI: 10.1038/s41598-024-77442-2] [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: 06/28/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
CRISPR-dead Cas9 interference (CRISPRi) has become a valuable tool for precise gene regulation. In this study, CRISPRi was designed to target the inhA gene of Mycobacterium smegmatis (Msm), a gene necessary for mycolic acid synthesis. Our findings revealed that sgRNA2 induced with 100 ng/ml aTc achieved over 90% downregulation of inhA gene expression and inhibited bacterial viability by approximately 1,000-fold. Furthermore, CRISPRi enhanced the susceptibility of M. smegmatis to isoniazid and rifampicin, which are both 50% and 90% lower than those of the wild-type strain or other strains, respectively. This study highlights the ability of CRISPRi to silence the inhA gene, which impacts bacterial viability and drug susceptibility. The findings provide valuable insights into the utility of CRISPRi as an alternative tool for gene regulation. CRISPRi might be further assessed for its synergistic effect with current anti-tuberculosis drugs and its possible implications for combating mycobacterial infections, especially drug-resistant tuberculosis.
Collapse
Affiliation(s)
- Nuntita Singpanomchai
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Panan Ratthawongjirakul
- Center of Excellence for Innovative Diagnosis of Antimicrobial Resistance, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
29
|
Bates NA, Rodriguez R, Drwich R, Ray A, Stanley SA, Penn BH. Reactive Oxygen Detoxification Contributes to Mycobacterium abscessus Antibiotic Survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618103. [PMID: 39554100 PMCID: PMC11565942 DOI: 10.1101/2024.10.13.618103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
When a population of bacteria encounter a bactericidal antibiotic most cells die rapidly. However, a sub-population, known as "persister cells", can survive for prolonged periods in a non-growing, but viable, state. Persister cell frequency is dramatically increased by stresses such as nutrient deprivation, but it is unclear what pathways are required to maintain viability, and how this process is regulated. To identify the genetic determinants of antibiotic persistence in mycobacteria, we carried out transposon mutagenesis high-throughput sequencing (Tn-Seq) screens in Mycobacterium abscessus (Mabs). This analysis identified genes essential in both spontaneous and stress-induced persister cells, allowing the first genetic comparison of these states in mycobacteria, and unexpectedly identified multiple genes involved in the detoxification of reactive oxygen species (ROS). We found that endogenous ROS were generated following antibiotic exposure, and that the KatG catalase-peroxidase contributed to survival in both spontaneous and starvation-induced persisters. We also found that that hypoxia significantly impaired bacterial killing, and notably, in the absence of oxygen, KatG became dispensable. Thus, the lethality of some antibiotics is amplified by toxic ROS accumulation, and persister cells depend on detoxification systems to remain viable.
Collapse
Affiliation(s)
- Nicholas A. Bates
- Department of Internal Medicine, University of California, Davis, California, USA
- Graduate Group in Immunology, University of California, Davis, California, USA
| | - Ronald Rodriguez
- Department of Molecular & Cell Biology, University of California, Berkeley, California, USA
- Department of Plant & Microbial Biology, University of California, Berkeley, California, USA
| | - Rama Drwich
- Department of Internal Medicine, University of California, Davis, California, USA
| | - Abigail Ray
- Microbiology Graduate Group, University of California, Davis, California, USA
| | - Sarah A. Stanley
- Department of Molecular & Cell Biology, University of California, Berkeley, California, USA
| | - Bennett H. Penn
- Department of Internal Medicine, University of California, Davis, California, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
30
|
Adolph C, Hards K, Williams ZC, Cheung CY, Keighley LM, Jowsey WJ, Kyte M, Inaoka DK, Kita K, Mackenzie JS, Steyn AJC, Li Z, Yan M, Tian GB, Zhang T, Ding X, Furkert DP, Brimble MA, Hickey AJR, McNeil MB, Cook GM. Identification of Chemical Scaffolds That Inhibit the Mycobacterium tuberculosis Respiratory Complex Succinate Dehydrogenase. ACS Infect Dis 2024; 10:3496-3515. [PMID: 39268963 DOI: 10.1021/acsinfecdis.3c00655] [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: 09/15/2024]
Abstract
Drug-resistant Mycobacterium tuberculosis is a significant cause of infectious disease morbidity and mortality for which new antimicrobials are urgently needed. Inhibitors of mycobacterial respiratory energy metabolism have emerged as promising next-generation antimicrobials, but a number of targets remain unexplored. Succinate dehydrogenase (SDH), a focal point in mycobacterial central carbon metabolism and respiratory energy production, is required for growth and survival in M. tuberculosis under a number of conditions, highlighting the potential of inhibitors targeting mycobacterial SDH enzymes. To advance SDH as a novel drug target in M. tuberculosis, we utilized a combination of biochemical screening and in-silico deep learning technologies to identify multiple chemical scaffolds capable of inhibiting mycobacterial SDH activity. Antimicrobial susceptibility assays show that lead inhibitors are bacteriostatic agents with activity against wild-type and drug-resistant strains of M. tuberculosis. Mode of action studies on lead compounds demonstrate that the specific inhibition of SDH activity dysregulates mycobacterial metabolism and respiration and results in the secretion of intracellular succinate. Interaction assays demonstrate that the chemical inhibition of SDH activity potentiates the activity of other bioenergetic inhibitors and prevents the emergence of resistance to a variety of drugs. Overall, this study shows that SDH inhibitors are promising next-generation antimicrobials against M. tuberculosis.
Collapse
Affiliation(s)
- Cara Adolph
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
| | - Kiel Hards
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
| | - Zoe C Williams
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Africa Health Research Institute, University of KwaZulu Natal, Durban 4001, South Africa
| | - Chen-Yi Cheung
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Laura M Keighley
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - William J Jowsey
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
| | - Matson Kyte
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Daniel Ken Inaoka
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
- Department of Host-Defence Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Jared S Mackenzie
- Africa Health Research Institute, University of KwaZulu Natal, Durban 4001, South Africa
| | - Adrie J C Steyn
- Africa Health Research Institute, University of KwaZulu Natal, Durban 4001, South Africa
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
- Centres for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Zhengqiu Li
- School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ming Yan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Guo-Bao Tian
- Department of Immunology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
- Advanced Medical Technology Centre, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
- Key Laboratory of Tropical Diseases Control, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaobo Ding
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
- School of Chemical Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Daniel P Furkert
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Margaret A Brimble
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
- School of Chemical Sciences, University of Auckland, Auckland 1010, New Zealand
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Anthony J R Hickey
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Matthew B McNeil
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
| | - Gregory M Cook
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1042, New Zealand
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| |
Collapse
|
31
|
Delbeau M, Froom R, Landick R, Darst SA, Campbell EA. The yin and yang of the universal transcription factor NusG. Curr Opin Microbiol 2024; 81:102540. [PMID: 39226817 PMCID: PMC11421859 DOI: 10.1016/j.mib.2024.102540] [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: 06/30/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
RNA polymerase (RNAP), the central enzyme of transcription, intermittently pauses during the elongation stage of RNA synthesis. Pausing provides an opportunity for regulatory events such as nascent RNA folding or the recruitment of transregulators. NusG (Spt5 in eukaryotes and archaea) regulates RNAP pausing and is the only transcription factor conserved across all cellular life. NusG is a multifunctional protein: its N-terminal domain (NGN) binds to RNAP, and its C-terminal KOW domain in bacteria interacts with transcription regulators such as ribosomes and termination factors. In Escherichia coli, NusG acts as an antipausing factor. However, recent studies have revealed that NusG has distinct transcriptional regulatory roles specific to bacterial clades with clinical implications. Here, we focus on NusG's dual roles in the regulation of pausing.
Collapse
Affiliation(s)
- Madeleine Delbeau
- Laboratory of Molecular Pathogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Ruby Froom
- Laboratory of Molecular Pathogenesis, The Rockefeller University, New York, NY 10065, USA
| | - Robert Landick
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Seth A Darst
- Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY 10065, USA
| | - Elizabeth A Campbell
- Laboratory of Molecular Pathogenesis, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
32
|
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
|
33
|
Yan M, Li H, Qu Y, Li S, Zheng D, Guo X, Wu Z, Lu J, Pang Y, Li W, Yang J, Zhan L, Sun Y. CRISPR Screening and Comparative LC-MS Analysis Identify Genes Mediating Efficacy of Delamanid and Pretomanid against Mycobacterium tuberculosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400176. [PMID: 39162029 PMCID: PMC11497083 DOI: 10.1002/advs.202400176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/23/2024] [Indexed: 08/21/2024]
Abstract
Tuberculosis (TB), the leading cause of death from bacterial infections worldwide, results from infection with Mycobacterium tuberculosis (Mtb). The antitubercular agents delamanid (DLM) and pretomanid (PMD) are nitroimidazole prodrugs that require activation by an enzyme intrinsic to Mtb; however, the mechanism(s) of action and the associated metabolic pathways are largely unclear. Profiling of the chemical-genetic interactions of PMD and DLM in Mtb using combined CRISPR screening reveals that the mutation of rv2073c increases susceptibility of Mtb to these nitroimidazole drugs both in vitro and in infected mice, whereas mutation of rv0078 increases drug resistance. Further assays show that Rv2073c might confer intrinsic resistance to DLM/PMD by interfering with inhibition of the drug target, decaprenylphophoryl-2-keto-b-D-erythro-pentose reductase (DprE2), by active nicotinamide adenine dinucleotide (NAD) adducts. Characterization of the metabolic pathways of DLM/PMD in Mtb using a combination of chemical genetics and comparative liquid chromatography-mass spectrometry (LC-MS) analysis of DLM/PMD metabolites reveals that Rv0077c, which is negatively regulated by Rv0078, mediates drug resistance by metabolizing activated DLM/PMD. These results might guide development of new nitroimidazole prodrugs and new regimens for TB treatment.
Collapse
Affiliation(s)
- Mei‐Yi Yan
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Haifeng Li
- NHC Key Laboratory of Human Disease Comparative Medicine, and National Center of Technology Innovation for Animal ModelInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021P. R. China
| | - Yun‐Mo Qu
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Si‐Shang Li
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Dandan Zheng
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Xiao‐Peng Guo
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Zhaojun Wu
- Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijing Chest HospitalCapital Medical UniversityBeijingP. R. China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck SurgeryBeijing Pediatric Research InstituteBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingP. R. China
| | - Yu Pang
- Department of Bacteriology and ImmunologyBeijing Chest HospitalCapital Medical UniversityBeijingP. R. China
| | - Weimin Li
- Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijing Chest HospitalCapital Medical UniversityBeijingP. R. China
| | - Jian Yang
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| | - Lingjun Zhan
- NHC Key Laboratory of Human Disease Comparative Medicine, and National Center of Technology Innovation for Animal ModelInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021P. R. China
| | - Yi‐Cheng Sun
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and MultimorbidityNational Institute of Pathogen Biology and Center for Tuberculosis ResearchChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100730P. R. China
| |
Collapse
|
34
|
Bustad E, Petry E, Gu O, Griebel BT, Rustad TR, Sherman DR, Yang JH, Ma S. Predicting bacterial fitness in Mycobacterium tuberculosis with transcriptional regulatory network-informed interpretable machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614645. [PMID: 39386570 PMCID: PMC11463588 DOI: 10.1101/2024.09.23.614645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis disease, the greatest source of global mortality by a bacterial pathogen. Mtb adapts and responds to diverse stresses such as antibiotics by inducing transcriptional stress-response regulatory programs. Understanding how and when these mycobacterial regulatory programs are activated could enable novel treatment strategies for potentiating the efficacy of new and existing drugs. Here we sought to define and analyze Mtb regulatory programs that modulate bacterial fitness. We assembled a large Mtb RNA expression compendium and applied these to infer a comprehensive Mtb transcriptional regulatory network and compute condition-specific transcription factor activity profiles. We utilized transcriptomic and functional genomics data to train an interpretable machine learning model that can predict Mtb fitness from transcription factor activity profiles. We demonstrated that this transcription factor activity-based model can successfully predict Mtb growth arrest and growth resumption under hypoxia and reaeration using only RNA-seq expression data as a starting point. These integrative network modeling and machine learning analyses thus enable the prediction of mycobacterial fitness under different environmental and genetic contexts. We envision these models can potentially inform the future design of prognostic assays and therapeutic intervention that can cripple Mtb growth and survival to cure tuberculosis disease.
Collapse
Affiliation(s)
- Ethan Bustad
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| | - Edson Petry
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Oliver Gu
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Braden T. Griebel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
| | | | - David R. Sherman
- Department of Microbiology, University of Washington, Seattle WA, USA
| | - Jason H. Yang
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
- Department of Microbiology, Biochemistry, & Molecular Genetics, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Shuyi Ma
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
- Department of Pediatrics, University of Washington, Seattle WA, USA
- Pathobiology Graduate Program, Department of Global Health, University of Washington, Seattle WA, USA
| |
Collapse
|
35
|
Eoh H, Lee JJ, Swanson D, Lee SK, Dihardjo S, Lee GY, Sree G, Maskill E, Taylor Z, Van Nieuwenhze M, Singh A, Lee JS, Eum SY, Cho SN, Swarts B. Trehalose catalytic shift is an intrinsic factor in Mycobacterium tuberculosis that enhances phenotypic heterogeneity and multidrug resistance. RESEARCH SQUARE 2024:rs.3.rs-4999164. [PMID: 39315249 PMCID: PMC11419184 DOI: 10.21203/rs.3.rs-4999164/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Drug-resistance (DR) in many bacterial pathogens often arises from the repetitive formation of drug-tolerant bacilli, known as persisters. However, it is unclear whether Mycobacterium tuberculosis (Mtb), the bacterium that causes tuberculosis (TB), undergoes a similar phenotypic transition. Recent metabolomics studies have identified that a change in trehalose metabolism is necessary for Mtb to develop persisters and plays a crucial role in metabolic networks of DR-TB strains. The present study used Mtb mutants lacking the trehalose catalytic shift and showed that the mutants exhibited a significantly lower frequency of the emergence of DR mutants compared to wildtype, due to reduced persister formation. The trehalose catalytic shift enables Mtb persisters to survive under bactericidal antibiotics by increasing metabolic heterogeneity and drug tolerance, ultimately leading to development of DR. Intriguingly, rifampicin (RIF)-resistant bacilli exhibit cross-resistance to a second antibiotic, due to a high trehalose catalytic shift activity. This phenomenon explains how the development of multidrug resistance (MDR) is facilitated by the acquisition of RIF resistance. In this context, the heightened risk of MDR-TB in the lineage 4 HN878 W-Beijing strain can be attributed to its greater trehalose catalytic shift. Genetic and pharmacological inactivation of the trehalose catalytic shift significantly reduced persister formation, subsequently decreasing the incidence of MDR-TB in HN878 W-Beijing strain. Collectively, the trehalose catalytic shift serves as an intrinsic factor of Mtb responsible for persister formation, cross-resistance to multiple antibiotics, and the emergence of MDR-TB. This study aids in the discovery of new TB therapeutics by targeting the trehalose catalytic shift of Mtb.
Collapse
|
36
|
Oluoch PO, Koh EI, Proulx MK, Reames CJ, Papavinasasundaram KG, Murphy KC, Zimmerman MD, Dartois V, Sassetti CM. Chemical genetic interactions elucidate pathways controlling tuberculosis antibiotic efficacy during infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.609063. [PMID: 39282290 PMCID: PMC11398305 DOI: 10.1101/2024.09.04.609063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Successful tuberculosis therapy requires treatment with an unwieldy multidrug combination for several months. Thus, there is a growing need to identify novel genetic vulnerabilities that can be leveraged to develop new, more effective antitubercular drugs. Consequently, recent efforts to optimize TB therapy have exploited Mtb chemical genetics to identify pathways influencing antibiotic efficacy, novel mechanisms of antibiotic action, and new targets for TB drug discovery. However, the influence of the complex host environment on these interactions remains largely unknown, leaving the therapeutic potential of the identified targets unclear. In this study, we leveraged a library of conditional mutants targeting 467 essential Mtb genes to characterize the chemical-genetic interactions (CGIs) with TB drugs directly in the mouse infection model. We found that these in vivo CGIs differ significantly from those identified in vitro . Both drug-specific and drug-agnostic effects were identified, and many were preserved during treatment with a multidrug combination, suggesting numerous strategies for enhancing therapy. This work also elucidated the complex effects of pyrazinamide (PZA), a drug that relies on aspects of the infection environment for efficacy. Specifically, our work supports the importance of coenzyme A synthesis inhibition during infection, as well as the antagonistic effect of iron limitation on PZA activity. In addition, we found that inhibition of thiamine and purine synthesis increases PZA efficacy, suggesting novel therapeutically exploitable metabolic dependencies. Our findings present a map of the unique in vivo CGIs, characterizing the mechanism of PZA activity in vivo and identifying novel targets for TB drug development. Significance The inevitable rise of multi-drug-resistant tuberculosis underscores the urgent need for new TB drugs and novel drug targets while prioritizing synergistic drug combinations. Chemical-genetic interaction (CGI) studies have delineated bacterial pathways influencing antibiotic efficacy and uncovered druggable pathways that synergize with TB drugs. However, most studies are conducted in vitro , limiting our understanding of how the host environment influences drug-mutant interactions. Using an inducible mutant library targeting essential Mtb genes to characterize CGIs during infection, this study reveals that CGIs are both drug-specific and drug-agnostic and differ significantly from those observed in vitro . Synergistic CGIs comprised distinct metabolic pathways mediating antibiotic efficacy, revealing novel drug mechanisms of action, and defining potential drug targets that would synergize with frontline antitubercular drugs.
Collapse
|
37
|
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
|
38
|
Trinidad DD, Macdonald CB, Rosenberg OS, Fraser JS, Coyote-Maestas W. Deep mutational scanning of EccD 3 reveals the molecular basis of its essentiality in the mycobacterium ESX secretion system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609456. [PMID: 39229178 PMCID: PMC11370616 DOI: 10.1101/2024.08.23.609456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Tuberculosis remains the deadliest infectious disease in the world and requires novel therapeutic targets. The ESX-3 secretion system, which is essential for iron and zinc homeostasis and thus M. tuberculosis survival, is a promising target. In this study, we perform a deep mutational scan on the ESX-3 core protein EccD3 in the model organism M. smegmatis. We systematically investigated the functional roles of 145 residues across the soluble ubiquitin-like domain, the conformationally distinct flexible linker, and selected transmembrane helices of EccD3. Our data combined with structural comparisons to ESX-5 complexes support a model where EccD3 stabilizes the complex, with the hinge motif within the linker being particularly sensitive to disruption. Our study is the first deep mutational scan in mycobacteria, which could help guide drug development toward novel treatment of tuberculosis. This study underscores the importance of context-specific mutational analyses for discovering essential protein interactions within mycobacterial systems.
Collapse
Affiliation(s)
| | - Christian B Macdonald
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Oren S Rosenberg
- Department of Medicine, University of California, San Francisco
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Willow Coyote-Maestas
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California, San Francisco, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
39
|
Chand P, Mendum TA, Butler RE, Hingley-Wilson SM, Stewart GR. Identification of gene targets that potentiate the action of rifampicin on Mycobacterium bovis BCG. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001488. [PMID: 39150447 PMCID: PMC11329110 DOI: 10.1099/mic.0.001488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Tuberculosis (TB) caused by bacteria of the Mycobacterium tuberculosis complex remains one of the most important infectious diseases of mankind. Rifampicin is a first line drug used in multi-drug treatment of TB, however, the necessary duration of treatment with these drugs is long and development of resistance is an increasing impediment to treatment programmes. As a result, there is a requirement for research and development of new TB drugs, which can form the basis of new drug combinations, either due to their own anti-mycobacterial activity or by augmenting the activity of existing drugs such as rifampicin. This study describes a TnSeq analysis to identify mutants with enhanced sensitivity to sub-minimum inhibitory concentrations (MIC) of rifampicin. The rifampicin-sensitive mutants were disrupted in genes of a variety of functions and the majority fitted into three thematic groups: firstly, genes that were involved in DNA/RNA metabolism, secondly, genes involved in sensing and regulating mycobacterial cellular systems, and thirdly, genes involved in the synthesis and maintenance of the cell wall. Selection at two concentrations of rifampicin (1/250 and 1/62 MIC) demonstrated a dose response for mutants with statistically significant sensitivity to rifampicin. The dataset reveals mechanisms of how mycobacteria are innately tolerant to and initiate an adaptive response to rifampicin; providing putative targets for the development of adjunctive therapies that potentiate the action of rifampicin.
Collapse
Affiliation(s)
- Pooja Chand
- Department of Microbial Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH United Kingdom
| | - Tom A. Mendum
- Department of Microbial Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH United Kingdom
| | - Rachel E. Butler
- Department of Microbial Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH United Kingdom
| | - Suzanne M. Hingley-Wilson
- Department of Microbial Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH United Kingdom
| | - Graham R. Stewart
- Department of Microbial Sciences, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH United Kingdom
| |
Collapse
|
40
|
Parkhill SL, Johnson EO. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochem J 2024; 481:839-864. [PMID: 38958473 PMCID: PMC11346456 DOI: 10.1042/bcj20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The application of dyes to understanding the aetiology of infection inspired antimicrobial chemotherapy and the first wave of antibacterial drugs. The second wave of antibacterial drug discovery was driven by rapid discovery of natural products, now making up 69% of current antibacterial drugs. But now with the most prevalent natural products already discovered, ∼107 new soil-dwelling bacterial species must be screened to discover one new class of natural product. Therefore, instead of a third wave of antibacterial drug discovery, there is now a discovery bottleneck. Unlike natural products which are curated by billions of years of microbial antagonism, the vast synthetic chemical space still requires artificial curation through the therapeutics science of antibacterial drugs - a systematic understanding of how small molecules interact with bacterial physiology, effect desired phenotypes, and benefit the host. Bacterial molecular genetics can elucidate pathogen biology relevant to therapeutics development, but it can also be applied directly to understanding mechanisms and liabilities of new chemical agents with new mechanisms of action. Therefore, the next phase of antibacterial drug discovery could be enabled by integrating chemical expertise with systematic dissection of bacterial infection biology. Facing the ambitious endeavour to find new molecules from nature or new-to-nature which cure bacterial infections, the capabilities furnished by modern chemical biology and molecular genetics can be applied to prospecting for chemical modulators of new targets which circumvent prevalent resistance mechanisms.
Collapse
Affiliation(s)
- Susannah L. Parkhill
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
| | - Eachan O. Johnson
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
- Department of Chemistry, Imperial College, London, U.K
- Department of Chemistry, King's College London, London, U.K
| |
Collapse
|
41
|
Miller AH, Marks F, Chan L, Botella H, Schnappinger D, Ehrt S. Interruption of mycothiol synthesis and intracellular redox status impact iron-regulated reporter activation in Mycobacterium smegmatis. Microbiol Spectr 2024; 12:e0048724. [PMID: 38860795 PMCID: PMC11218476 DOI: 10.1128/spectrum.00487-24] [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: 05/04/2024] [Indexed: 06/12/2024] Open
Abstract
Iron scavenging is required for full virulence of mycobacterial pathogens. During infection, the host immune response restricts mycobacterial access to iron, which is essential for bacterial respiration and DNA synthesis. The Mycobacterium tuberculosis iron-dependent regulator (IdeR) responds to changes in iron accessibility by repressing iron-uptake genes when iron is available. In contrast, iron-uptake gene transcription is induced when iron is depleted. The ideR gene is essential in M. tuberculosis and is required for bacterial growth. To further study how iron regulates transcription, wee developed an iron responsive reporter system that relies on an IdeR-regulated promoter to drive Cre and loxP mediated recombination in Mycobacterium smegmatis. Recombination leads to the expression of an antibiotic resistance gene so that mutations that activate the IdeR-regulated promoter can be selected. A transposon library in the background of this reporter system was exposed to media containing iron and hemin, and this resulted in the selection of mutants in the antioxidant mycothiol synthesis pathway. We validated that inactivation of the mycothiol synthesis gene mshA results in increased recombination and increased IdeR-regulated promoter activity in the reporter system. Further, we show that vitamin C, which has been shown to oxidize iron through the Fenton reaction, can decrease promoter activity in the mshA mutant. We conclude that the intracellular redox state balanced by mycothiol can alter IdeR activity in the presence of iron.IMPORTANCEMycobacterium smegmatis is a tractable organism to study mycobacterial gene regulation. We used M. smegmatis to construct a novel recombination-based reporter system that allows for the selection of mutations that deregulate a promoter of interest. Transposon mutagenesis and insertion sequencing (TnSeq) in the recombination reporter strain identified genes that impact iron regulated promoter activity in mycobacteria. We found that the mycothiol synthesis gene mshA is required for IdeR mediated transcriptional regulation by maintaining intracellular redox balance. By affecting the oxidative state of the intracellular environment, mycothiol can modulate iron-dependent transcriptional activity. Taken more broadly, this novel reporter system can be used in combination with transposon mutagenesis to identify genes that are required by Mycobacterium tuberculosis to overcome temporary or local changes in iron availability during infection.
Collapse
Affiliation(s)
- Alexandra H Miller
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York, USA
| | - Frances Marks
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
- Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York, USA
| | - Luming Chan
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Helene Botella
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Dirk Schnappinger
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
| | - Sabine Ehrt
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, New York, USA
| |
Collapse
|
42
|
Habjan E, Lepioshkin A, Charitou V, Egorova A, Kazakova E, Ho VQ, Bitter W, Makarov V, Speer A. Modulating mycobacterial envelope integrity for antibiotic synergy with benzothiazoles. Life Sci Alliance 2024; 7:e202302509. [PMID: 38744470 PMCID: PMC11094368 DOI: 10.26508/lsa.202302509] [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: 12/04/2023] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Developing effective tuberculosis drugs is hindered by mycobacteria's intrinsic antibiotic resistance because of their impermeable cell envelope. Using benzothiazole compounds, we aimed to increase mycobacterial cell envelope permeability and weaken the defenses of Mycobacterium marinum, serving as a model for Mycobacterium tuberculosis Initial hit, BT-08, significantly boosted ethidium bromide uptake, indicating enhanced membrane permeability. It also demonstrated efficacy in the M. marinum-zebrafish embryo infection model and M. tuberculosis-infected macrophages. Notably, BT-08 synergized with established antibiotics, including vancomycin and rifampicin. Subsequent medicinal chemistry optimization led to BT-37, a non-toxic and more potent derivative, also enhancing ethidium bromide uptake and maintaining synergy with rifampicin in infected zebrafish embryos. Mutants of M. marinum resistant to BT-37 revealed that MMAR_0407 (Rv0164) is the molecular target and that this target plays a role in the observed synergy and permeability. This study introduces novel compounds targeting a new mycobacterial vulnerability and highlights their cooperative and synergistic interactions with existing antibiotics.
Collapse
Affiliation(s)
- Eva Habjan
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Location VU Medical Center, Amsterdam, Netherlands
| | - Alexander Lepioshkin
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Vicky Charitou
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Location VU Medical Center, Amsterdam, Netherlands
| | - Anna Egorova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Elena Kazakova
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Vien Qt Ho
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Location VU Medical Center, Amsterdam, Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Location VU Medical Center, Amsterdam, Netherlands
| | - Vadim Makarov
- Federal Research Centre "Fundamentals of Biotechnology" of the Russian Academy of Sciences (Research Centre of Biotechnology RAS), Moscow, Russia
| | - Alexander Speer
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Location VU Medical Center, Amsterdam, Netherlands
| |
Collapse
|
43
|
Sengupta S, Jain P, Ratnam R, Pandey BK, Singh U, Kumar V, Paliwal A, Jain A. Agreement between Phenotypically Detected Linezolid Resistance and Mutations in rrl and rplC Genes of Mycobacterium tuberculosis Isolates Using Nanopore Sequencing. Int J Mycobacteriol 2024; 13:331-336. [PMID: 39277897 DOI: 10.4103/ijmy.ijmy_139_24] [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: 05/12/2024] [Accepted: 08/19/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Phenotypic drug susceptibility testing (DST) is considered the gold standard for detecting linezolid (LZD) resistance in Mycobacterium tuberculosis (MTB), but it is time-consuming. Nanopore sequencing offers a potentially faster alternative approach. This study evaluated the agreement between phenotypically detected LZD resistance and mutations in the rrl and rplC genes of MTB isolates using nanopore sequencing. METHODS Consecutive drug-resistant MTB isolates from pulmonary samples collected in 2021 underwent liquid culture (LC) DST for LZD. All resistant isolates and an equal number of susceptible isolates were subjected to targeted sequencing of the rrl and rplC genes using nanopore technology. RESULTS Sequencing identified a C154R mutation in the rplC gene in only one LZD-resistant isolate. No mutations were detected in the rrl gene. The agreement between sequencing and LC-DST for detecting LZD resistance was poor (Cohen's kappa: 0.03571, 95% confidence interval [CI]: -0.034-0.105). Additionally, no significant association was found between LZD resistance and clinical or microbiological outcomes at 6-month follow-up. CONCLUSION This study revealed a considerable discrepancy between phenotypic and genotypic detection of LZD resistance in MTB. Further research is needed to better understand the genetic mechanisms underlying LZD resistance and to develop reliable molecular diagnostics for rapid resistance detection.
Collapse
|
44
|
Rahman ASMZ, Syroegin EA, Novomisky Nechcoff J, Devarajan A, Polikanov YS, Cardona ST. Rationally Designed Pooled CRISPRi-Seq Uncovers an Inhibitor of Bacterial Peptidyl-tRNA Hydrolase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592284. [PMID: 38979387 PMCID: PMC11230203 DOI: 10.1101/2024.05.02.592284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pooled knockdown libraries of essential genes are useful tools for elucidating the mechanisms of action of antibacterial compounds, a pivotal step in antibiotic discovery. However, achieving genomic coverage of antibacterial targets poses a challenge due to the uneven proliferation of knockdown mutants during pooled growth, leading to the unintended loss of important targets. To overcome this issue, we describe the construction of CIMPLE ( C RISPR i - m ediated p ooled library of e ssential genes), a rationally designed pooled knockdown library built in a model antibiotic-resistant bacteria, Burkholderia cenocepacia. By analyzing growth parameters of clonal knockdown populations of an arrayed CRISPRi library, we predicted strain depletion levels during pooled growth and adjusted mutant relative abundance, approaching genomic coverage of antibacterial targets during antibiotic exposure. We first benchmarked CIMPLE by chemical-genetic profiling of known antibacterials, then applied it to an uncharacterized bacterial growth inhibitor from a new class. CRISPRi-Seq with CIMPLE, followed by biochemical validation, revealed that the novel compound targets the peptidyl-tRNA hydrolase (Pth). Overall, CIMPLE leverages the advantages of arrayed and pooled CRISPRi libraries to uncover unexplored targets for antibiotic action. Summary Bacterial mutant libraries in which antibiotic targets are downregulated are useful tools to functionally characterize novel antimicrobials. These libraries are used for chemical-genetic profiling as target-compound interactions can be inferred by differential fitness of mutants during pooled growth. Mutants that are functionally related to the antimicrobial mode of action are usually depleted from the pool upon exposure to the drug. Although powerful, this method can fail when the unequal proliferation of mutant strains before exposure causes mutants to fall below the detection level in the library pool. To address this issue, we constructed an arrayed essential gene mutant library (EGML) in the antibiotic-resistant bacterium Burkholderia cenocepacia using CRISPR interference (CRISPRi) and analyzed the growth parameters of individual mutant strains. We then modelled depletion levels during pooled growth and used the model to rationally design an optimized CRISPR interference-mediated pooled library of essential genes (CIMPLE). By adjusting the initial inoculum of the knockdown mutants, we achieved coverage of the bacterial essential genome with mutant sensitization. We exposed CIMPLE to a recently discovered antimicrobial of a novel class and discovered it inhibits the peptidyl-tRNA hydrolase, an essential bacterial enzyme. In summary, we demonstrate the utility of CIMPLE and CRISPRi-Seq to uncover the mechanism of action of novel antimicrobial compounds. Graphical abstract
Collapse
|
45
|
Enright AL, Heelan WJ, Ward RD, Peters JM. CRISPRi functional genomics in bacteria and its application to medical and industrial research. Microbiol Mol Biol Rev 2024; 88:e0017022. [PMID: 38809084 PMCID: PMC11332340 DOI: 10.1128/mmbr.00170-22] [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] [Indexed: 05/30/2024] Open
Abstract
SUMMARYFunctional genomics is the use of systematic gene perturbation approaches to determine the contributions of genes under conditions of interest. Although functional genomic strategies have been used in bacteria for decades, recent studies have taken advantage of CRISPR (clustered regularly interspaced short palindromic repeats) technologies, such as CRISPRi (CRISPR interference), that are capable of precisely modulating expression of all genes in the genome. Here, we discuss and review the use of CRISPRi and related technologies for bacterial functional genomics. We discuss the strengths and weaknesses of CRISPRi as well as design considerations for CRISPRi genetic screens. We also review examples of how CRISPRi screens have defined relevant genetic targets for medical and industrial applications. Finally, we outline a few of the many possible directions that could be pursued using CRISPR-based functional genomics in bacteria. Our view is that the most exciting screens and discoveries are yet to come.
Collapse
Affiliation(s)
- Amy L. Enright
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- DOE Great Lakes Bioenergy Research Center University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - William J. Heelan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ryan D. Ward
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- DOE Great Lakes Bioenergy Research Center University of Wisconsin-Madison, Madison, Wisconsin, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jason M. Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- DOE Great Lakes Bioenergy Research Center University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
46
|
Romano KP, Bagnall J, Warrier T, Sullivan J, Ferrara K, Orzechowski M, Nguyen P, Raines K, Livny J, Shoresh N, Hung D. Perturbation-Specific Transcriptional Mapping for unbiased target elucidation of antibiotics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.590978. [PMID: 38712067 PMCID: PMC11071498 DOI: 10.1101/2024.04.25.590978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The rising prevalence of antibiotic resistance threatens human health. While more sophisticated strategies for antibiotic discovery are being developed, target elucidation of new chemical entities remains challenging. In the post-genomic era, expression profiling can play an important role in mechanism-of-action (MOA) prediction by reporting on the cellular response to perturbation. However, the broad application of transcriptomics has yet to fulfill its promise of transforming target elucidation due to challenges in identifying the most relevant, direct responses to target inhibition. We developed an unbiased strategy for MOA prediction, called Perturbation-Specific Transcriptional Mapping (PerSpecTM), in which large-throughput expression profiling of wildtype or hypomorphic mutants, depleted for essential targets, enables a computational strategy to address this challenge. We applied PerSpecTM to perform reference-based MOA prediction based on the principle that similar perturbations, whether chemical or genetic, will elicit similar transcriptional responses. Using this approach, we elucidated the MOAs of three new molecules with activity against Pseudomonas aeruginosa by comparing their expression profiles to those of a reference set of antimicrobial compounds with known MOAs. We also show that transcriptional responses to small molecule inhibition resemble those resulting from genetic depletion of essential targets by CRISPRi by PerSpecTM, demonstrating proof-of-concept that correlations between expression profiles of small molecule and genetic perturbations can facilitate MOA prediction when no chemical entities exist to serve as a reference. Empowered by PerSpecTM, this work lays the foundation for an unbiased, readily scalable, systematic reference-based strategy for MOA elucidation that could transform antibiotic discovery efforts.
Collapse
|
47
|
Negatu DA, Aragaw WW, Dartois V, Dick T. A pairwise approach to revitalize β-lactams for the treatment of TB. Antimicrob Agents Chemother 2024; 68:e0003424. [PMID: 38690896 PMCID: PMC11620507 DOI: 10.1128/aac.00034-24] [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: 01/05/2024] [Accepted: 04/07/2024] [Indexed: 05/03/2024] Open
Abstract
The dual β-lactam approach has been successfully applied to overcome target redundancy in nontuberculous mycobacteria. Surprisingly, this approach has not been leveraged for Mycobacterium tuberculosis, despite the high conservation of peptidoglycan synthesis. Through a comprehensive screen of oral β-lactam pairs, we have discovered that cefuroxime strongly potentiates the bactericidal activity of tebipenem and sulopenem-advanced clinical candidates-and amoxicillin, at concentrations achieved clinically. β-lactam pairs thus have the potential to reduce TB treatment duration.
Collapse
Affiliation(s)
- Dereje A. Negatu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Center for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), Addis Ababa University, Addis Ababa, Ethiopia
| | - Wassihun Wedajo Aragaw
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
48
|
Mulholland CV, Wiggins TJ, Cui J, Vilchèze C, Rajagopalan S, Shultis MW, Reyes-Fernández EZ, Jacobs WR, Berney M. Propionate prevents loss of the PDIM virulence lipid in Mycobacterium tuberculosis. Nat Microbiol 2024; 9:1607-1618. [PMID: 38740932 PMCID: PMC11253637 DOI: 10.1038/s41564-024-01697-8] [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: 10/13/2023] [Accepted: 04/04/2024] [Indexed: 05/16/2024]
Abstract
Phthiocerol dimycocerosate (PDIM) is an essential virulence lipid of Mycobacterium tuberculosis. In vitro culturing rapidly selects for spontaneous PDIM-negative mutants that have attenuated virulence and increased cell wall permeability, thus impacting the relevance of experimental findings. PDIM loss can also reduce the efficacy of the BCG Pasteur vaccine. Here we show that vancomycin susceptibility can rapidly screen for M. tuberculosis PDIM production. We find that metabolic deficiency of methylmalonyl-CoA impedes the growth of PDIM-producing bacilli, selecting for PDIM-negative variants. Supplementation with odd-chain fatty acids, cholesterol or vitamin B12 restores PDIM-positive bacterial growth. Specifically, we show that propionate supplementation enhances PDIM-producing bacterial growth and selects against PDIM-negative mutants, analogous to in vivo conditions. Our study provides a simple approach to screen for and maintain PDIM production, and reveals how discrepancies between the host and in vitro nutrient environments can attenuate bacterial pathogenicity.
Collapse
Affiliation(s)
- Claire V Mulholland
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Thomas J Wiggins
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Jinhua Cui
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Catherine Vilchèze
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Saranathan Rajagopalan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Michael W Shultis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | | | - William R Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Michael Berney
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
49
|
Stanley S, Spaulding CN, Liu Q, Chase MR, Ha DTM, Thai PVK, Lan NH, Thu DDA, Quang NL, Brown J, Hicks ND, Wang X, Marin M, Howard NC, Vickers AJ, Karpinski WM, Chao MC, Farhat MR, Caws M, Dunstan SJ, Thuong NTT, Fortune SM. Identification of bacterial determinants of tuberculosis infection and treatment outcomes: a phenogenomic analysis of clinical strains. THE LANCET. MICROBE 2024; 5:e570-e580. [PMID: 38734030 PMCID: PMC11229950 DOI: 10.1016/s2666-5247(24)00022-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 12/23/2023] [Accepted: 01/16/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Bacterial diversity could contribute to the diversity of tuberculosis infection and treatment outcomes observed clinically, but the biological basis of this association is poorly understood. The aim of this study was to identify associations between phenogenomic variation in Mycobacterium tuberculosis and tuberculosis clinical features. METHODS We developed a high-throughput platform to define phenotype-genotype relationships in M tuberculosis clinical isolates, which we tested on a set of 158 drug-sensitive M tuberculosis strains sampled from a large tuberculosis clinical study in Ho Chi Minh City, Viet Nam. We tagged the strains with unique genetic barcodes in multiplicate, allowing us to pool the strains for in-vitro competitive fitness assays across 16 host-relevant antibiotic and metabolic conditions. Relative fitness was quantified by deep sequencing, enumerating output barcode read counts relative to input normalised values. We performed a genome-wide association study to identify phylogenetically linked and monogenic mutations associated with the in-vitro fitness phenotypes. These genetic determinants were further associated with relevant clinical outcomes (cavitary disease and treatment failure) by calculating odds ratios (ORs) with binomial logistic regressions. We also assessed the population-level transmission of strains associated with cavitary disease and treatment failure using terminal branch length analysis of the phylogenetic data. FINDINGS M tuberculosis clinical strains had diverse growth characteristics in host-like metabolic and drug conditions. These fitness phenotypes were highly heritable, and we identified monogenic and phylogenetically linked variants associated with the fitness phenotypes. These data enabled us to define two genetic features that were associated with clinical outcomes. First, mutations in Rv1339, a phosphodiesterase, which were associated with slow growth in glycerol, were further associated with treatment failure (OR 5·34, 95% CI 1·21-23·58, p=0·027). Second, we identified a phenotypically distinct slow-growing subclade of lineage 1 strains (L1.1.1.1) that was associated with cavitary disease (OR 2·49, 1·11-5·59, p=0·027) and treatment failure (OR 4·76, 1·53-14·78, p=0·0069), and which had shorter terminal branch lengths on the phylogenetic tree, suggesting increased transmission. INTERPRETATION Slow growth under various antibiotic and metabolic conditions served as in-vitro intermediate phenotypes underlying the association between M tuberculosis monogenic and phylogenetically linked mutations and outcomes such as cavitary disease, treatment failure, and transmission potential. These data suggest that M tuberculosis growth regulation is an adaptive advantage for bacterial success in human populations, at least in some circumstances. These data further suggest markers for the underlying bacterial processes that contribute to these clinical outcomes. FUNDING National Health and Medical Research Council/A∗STAR, National Institutes of Allergy and Infectious Diseases, National Institute of Child Health and Human Development, and the Wellcome Trust Fellowship in Public Health and Tropical Medicine.
Collapse
Affiliation(s)
- Sydney Stanley
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Caitlin N Spaulding
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Qingyun Liu
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Michael R Chase
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | | | | | | | - Do Dang Anh Thu
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Nguyen Le Quang
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam
| | - Jessica Brown
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Nathan D Hicks
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Xin Wang
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Maximillian Marin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nicole C Howard
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Andrew J Vickers
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Wiktor M Karpinski
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Michael C Chao
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Maha R Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Maxine Caws
- Liverpool School of Tropical Medicine, Liverpool, UK; Birat Nepal Medical Trust, Kathmandu, Nepal
| | - Sarah J Dunstan
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Nguyen Thuy Thuong Thuong
- Oxford University Clinical Research Unit, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam; Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T H Chan School of Public Health, Boston, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
50
|
Jowsey WJ, Cook GM, McNeil MB. Antibiotic resistance in Mycobacterium tuberculosis alters tolerance to cell wall-targeting inhibitors. JAC Antimicrob Resist 2024; 6:dlae086. [PMID: 38836195 PMCID: PMC11148391 DOI: 10.1093/jacamr/dlae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/11/2024] [Indexed: 06/06/2024] Open
Abstract
Background A limited ability to eliminate drug-resistant strains of Mycobacterium tuberculosis is a major contributor to the morbidity of TB. Complicating this problem, little is known about how drug resistance-conferring mutations alter the ability of M. tuberculosis to tolerate antibiotic killing. Here, we investigated if drug-resistant strains of M. tuberculosis have an altered ability to tolerate killing by cell wall-targeting inhibitors. Methods Bacterial killing and MIC assays were used to test for antibiotic tolerance and synergy against a panel of drug-resistant M. tuberculosis strains. Results Our results demonstrate that vancomycin and thioacetazone exhibit increased killing of diverse drug-resistant strains. Mutations in mmaA4 and mmpL3 increased vancomycin killing, which was consistent with vancomycin synergizing with thioacetazone and MmpL3-targeting inhibitors. In contrast, mutations in the mce1 operon conferred tolerance to vancomycin. Conclusions Overall, this work demonstrates how drug-resistant strains experience perturbations in cell-wall production that alters their tolerance to killing by cell wall-targeting inhibitors.
Collapse
Affiliation(s)
- William J Jowsey
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Gregory M Cook
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matthew B McNeil
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|