1
|
Li J, Liu D, Li X, Wei J, Du W, Zhao A, Xu M. RNA vaccines: The dawn of a new age for tuberculosis? Hum Vaccin Immunother 2025; 21:2469333. [PMID: 40013818 PMCID: PMC11869779 DOI: 10.1080/21645515.2025.2469333] [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: 12/26/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025] Open
Abstract
Since 2019, there has been a growing focus on mRNA vaccines for infectious disease prevention, particularly following the emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). mRNA vaccines offer advantages such as rapid production and the ability to induce robust cellular and antibody responses, which are essential for combating infections that require cell-mediated immunity, including Tuberculosis (TB). This review explores recent progress in TB mRNA vaccines and addresses several key areas: (1) the urgent need for new TB vaccines; (2) current advancements in TB vaccine development, and the advantages and challenges of mRNA technology; (3) the design and characteristics of TB mRNA vaccines; (4) the immunological mechanisms of TB mRNA vaccines; (5) manufacturing processes for TB mRNA vaccines; and (6) safety and regulatory considerations. This interdisciplinary review aims to provide insights for researchers working to address critical questions in TB mRNA vaccine development.
Collapse
Affiliation(s)
- Junli Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Dong Liu
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Xiaochi Li
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Jiazheng Wei
- College of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Weixin Du
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Aihua Zhao
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| | - Miao Xu
- Division of Tuberculosis Vaccine and Allergen Products, Institute of Biological Product Control, National Institutes for Food and Drug Control, Beijing, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
- Key Laboratory for Quality Research and Evaluation of Biological Products, National Medical Products Administration (NMPA), Beijing, China
- Key Laboratory of Research on Quality and Standardization of Biotech Products, National Health Commission (NHC), Beijing, China
| |
Collapse
|
2
|
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] [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
|
3
|
Liu P, Deng J, Yang Y, Bai W, Dong S, Zhang Z. Mycobacterium tuberculosis specific protein Rv1509 modulates osteoblast and osteoclast differentiation via TLR2 signaling. iScience 2025; 28:112107. [PMID: 40129707 PMCID: PMC11931388 DOI: 10.1016/j.isci.2025.112107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/09/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb), is one of the most ancient diseases recorded. In cases of bone TB, it significantly disrupts bone homeostasis, though the precise mechanisms are poorly understood and effective treatment targets are scarce. Our study investigated the role of Rv1509 in the pathogenesis of bone TB. We found that Rv1509 enhances the differentiation of bone marrow macrophages (BMMs) into osteoclasts by activating the TLR2 pathway, which stimulates the production of IL-6 and TNF-α. This, in turn, indirectly inhibits osteoblast differentiation and mineralization. Additionally, Rv1509 directly impairs osteoblast function and enhances the secretion of RANKL via TLR2 signaling, creating a detrimental RANKL/OPG imbalance that promotes osteoclast differentiation and bone degradation. Notably, the injection of Rv1509 into mouse skulls led to extensive bone damage, highlighting its significant role as a virulence factor in the pathogenesis of bone TB.
Collapse
Affiliation(s)
- Pan Liu
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Jiezhong Deng
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Yusheng Yang
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Wenxi Bai
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Shengtao Dong
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Zehua Zhang
- Institute of Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing 400038, China
| |
Collapse
|
4
|
Recchia D, Stelitano G, Egorova A, Batisti Biffignandi G, Savková K, Kafková R, Huszár S, Marino Cerrato A, Slayden RA, Cummings JE, Whittel N, Bauman AA, Robertson GT, Rank L, Urbina F, Lane TR, Ekins S, Riabova O, Kazakova E, Mikušová K, Sassera D, Degiacomi G, Chiarelli LR, Makarov V, Pasca MR. Mycobacterium tuberculosis Sulfate Ester Dioxygenase Rv3406 Is Able to Inactivate the RCB18350 Compound. ACS Infect Dis 2025. [PMID: 40111403 DOI: 10.1021/acsinfecdis.4c01030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Among the critical priority pathogens listed by the World Health Organization, Mycobacterium tuberculosis strains resistant to rifampicin present a significant global threat. Consequently, the study of the mechanisms of resistance to new antitubercular drugs and the discovery of new effective molecules are two crucial points in tuberculosis drug discovery. In this study, we discovered a compound named RCB18350, which is active against M. tuberculosis growth and exhibits a minimum inhibitory concentration (MIC) of 1.25 μg/mL. It was also effective against multidrug-resistant isolates. We deeply studied the mechanism of resistance/action of RCB18350 by using several approaches. We found that Rv3406, an iron- and α-ketoglutarate-dependent sulfate ester dioxygenase, is capable of metabolizing the compound into its inactive metabolite. This finding highlights the role of this enzyme in the mechanism of resistance to RCB18350.
Collapse
Affiliation(s)
- Deborah Recchia
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
| | - Giovanni Stelitano
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
| | - Anna Egorova
- Research Centre of Biotechnology RAS, Moscow 119071, Russia
| | | | - Karin Savková
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 814 99 Bratislava, Slovakia
| | - Radka Kafková
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 814 99 Bratislava, Slovakia
| | - Stanislav Huszár
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 814 99 Bratislava, Slovakia
| | - Antonio Marino Cerrato
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
| | - Richard A Slayden
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Jason E Cummings
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Nicholas Whittel
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Allison A Bauman
- Colorado State University, 1682 Campus Delivery, 200 West Lake Street, Fort Collins, Colorado 80523-1782, United States
| | - Gregory T Robertson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Laura Rank
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Research Centre of Biotechnology RAS, Moscow 119071, Russia
| | - Elena Kazakova
- Research Centre of Biotechnology RAS, Moscow 119071, Russia
| | - Katarína Mikušová
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, 814 99 Bratislava, Slovakia
| | - Davide Sassera
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
- Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giulia Degiacomi
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
| | - Laurent Robert Chiarelli
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
| | - Vadim Makarov
- Research Centre of Biotechnology RAS, Moscow 119071, Russia
| | - Maria Rosalia Pasca
- Department of Biology and Biotechnology "Lazzaro Spallanzani,", University of Pavia, 27100 Pavia, Italy
- Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
5
|
He X, He Y, Deng X, Lu N, Li A, Gao S, He S, Wang Y, Fu N, Wang Z, Nie Y, Xu L. Rv2741 Promotes Mycobacterium Survival by Modulating Macrophage Function via the IL-1α-MAPK Axis. ACS Infect Dis 2025; 11:676-688. [PMID: 40009799 DOI: 10.1021/acsinfecdis.4c00790] [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: 02/28/2025]
Abstract
One of the primary healthcare problems in the world today is tuberculosis (TB), a chronic infectious illness brought on by Mycobacterium tuberculosis (M. tuberculosis). A distinct family of PE_PGRS proteins, encoded by the M. tuberculosis genome, has attracted more attention because of their involvement in immune evasion and bacterial pathogenicity. Nevertheless, the specific functions and mechanisms of action for the majority of PE_PGRS proteins remain largely unexplored. This study focuses on the Rv2741 (PE_PGRS47) gene, which is exclusively present in pathogenic mycobacteria. To examine the function of Rv2741 in host-pathogen interactions, we created recombinant strains of Mycobacterium smegmatis (M. smegmatis) that expressed the M. tuberculosis Rv2741 gene. IL-1α was found to be a key mediator of host response modulation by Rv2741. Rv2741 downregulates the secretion of IL-1α and inhibits the MAPK signaling pathway, particularly the p38 and ERK1/2 pathways, thereby cooperatively inhibiting macrophage autophagy and apoptosis. Meanwhile, the decrease in IL-1α secretion directly leads to changes in the cytokine secretion pattern and a reduction in nitric oxide (NO) production. This multifaceted regulatory mechanism ultimately favors the survival of M. smegmatis in macrophages. This research significantly expands our understanding of Rv2741 function, revealing its crucial role as a multifunctional virulence factor in the immune evasion of M. tuberculosis.
Collapse
Affiliation(s)
- Xintong He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yonglin He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xichuan Deng
- Pathogen Biology and Immunology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 400016, China
| | - Nan Lu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Anlong Li
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Sijia Gao
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shiyan He
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuran Wang
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Nanzhe Fu
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Zijie Wang
- International Medical School, Chongqing Medical University, Chongqing 400016, China
| | - Yuxin Nie
- The Second Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Lei Xu
- Department of Pathogenic Biology, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
6
|
Mishra S, Singh PR, Hu X, Lopez-Quezada L, Jinich A, Jahn R, Geurts L, Shen N, DeJesus MA, Hartman T, Rhee K, Zimmerman M, Dartois V, Jones RM, Jiang X, Almada-Monter R, Bourouiba L, Nathan C. Candidate transmission survival genome of Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2025; 122:e2425981122. [PMID: 40053362 PMCID: PMC11912377 DOI: 10.1073/pnas.2425981122] [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: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 03/19/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb), a leading cause of death from infection, completes its life cycle entirely in humans except for transmission through the air. To begin to understand how Mtb survives aerosolization, we mimicked liquid and atmospheric conditions experienced by Mtb before and after exhalation using a model aerosol fluid (MAF) based on the water-soluble, lipidic, and cellular constituents of necrotic tuberculosis lesions. MAF induced drug tolerance in Mtb, remodeled its transcriptome, and protected Mtb from dying in microdroplets desiccating in air. Yet survival was not passive: Mtb appeared to rely on hundreds of genes to survive conditions associated with transmission. Essential genes subserving proteostasis offered most protection. A large number of conventionally nonessential genes appeared to contribute as well, including genes encoding proteins that resemble antidesiccants. The candidate transmission survival genome of Mtb may offer opportunities to reduce transmission of tuberculosis.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Prabhat Ranjan Singh
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Xiaoyi Hu
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Landys Lopez-Quezada
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Adrian Jinich
- Department of Chemistry and Biochemistry, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA92093-0021
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA92093-0021
| | - Robin Jahn
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Luc Geurts
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Naijian Shen
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Michael A. DeJesus
- Laboratory of Host-Pathogen Biology, Rockefeller University, New York, NY10021
| | - Travis Hartman
- Department of Medicine, Weill Cornell Medicine, New York, NY10065
| | - Kyu Rhee
- Department of Medicine, Weill Cornell Medicine, New York, NY10065
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Veronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Richard M. Jones
- Department of Microbiology, University of Washington, Seattle, WA98195
| | - Xiuju Jiang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Ricardo Almada-Monter
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA92093-0021
| | - Lydia Bourouiba
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
7
|
Pacce V, Guimarães AM, Kremer FS, Ferreira GN, Vedova-Costa JMD, dos Santos AC, Dellagostin OA, Soccol CR, Thomaz-Soccol V. Integrated Bioinformatics Analysis for Target Identification and Evaluation of Recombinant Protein as an Antigen for Intradermal Skin Test in Bovine Tuberculosis Diagnosis. ACS OMEGA 2025; 10:9187-9196. [PMID: 40092765 PMCID: PMC11904847 DOI: 10.1021/acsomega.4c09374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Bovine tuberculosis (bTB) is a respiratory disease caused by Mycobacterium bovis, posing a significant threat to animal health and the livestock industry. Current control strategies for bTB rely on diagnostic tests and slaughter policies. However, the limitations of existing diagnostic methods, which depend on PPD antigens, necessitate the exploration of alternative antigens to enhance the accuracy and reliability of bTB diagnosis. This study aimed to identify, produce, and evaluate novel antigens for use in the intradermal skin test for bTB diagnosis. A pangenome analysis of four Mycobacterium species identified 12 unique genes specific to M. bovis SP38. Further integrated bioinformatic analysis revealed 224 genomic islands associated with virulence and pathogenesis. Among these, a highly antigenic protein, termed HP28, was selected for in vivo testing. The recombinant HP28 protein (rHP28) was expressed in E. coli and assessed for its ability to induce intradermal skin reactions in guinea pigs. The rHP28 protein elicited a skin reaction of 6.6 mm at 72 h post-injection, whereas negative controls showed no reaction. This study presents a pipeline for the selection of antigens using integrated bioinformatic analysis to identify diagnostic targets that can effectively distinguish between sensitized and non-sensitized animals, offering a promising approach for improving bTB diagnostics.
Collapse
Affiliation(s)
- Violetta
Dias Pacce
- Laboratório
de Biologia Molecular, Programa de Pós Graduação
em Engenharia de Bioprocessos e Biotecnologia, Universidade Federal do Paraná, Curitiba, Paraná 81531-990, Brazil
| | - Amanda Munari Guimarães
- Programa
de Pós Graduação em Biotecnologia, Centro de
Desenvolvimento Tecnológico, Universidade
Federal de Pelotas, Pelotas, Rio Grande do Sul 96160-000, Brazil
| | - Frederico Schmitt Kremer
- Programa
de Pós Graduação em Biotecnologia, Centro de
Desenvolvimento Tecnológico, Universidade
Federal de Pelotas, Pelotas, Rio Grande do Sul 96160-000, Brazil
| | - Gabriela Nascimento Ferreira
- Laboratório
de Biologia Molecular, Programa de Pós Graduação
em Engenharia de Bioprocessos e Biotecnologia, Universidade Federal do Paraná, Curitiba, Paraná 81531-990, Brazil
| | - Jean Michel Dela Vedova-Costa
- Laboratório
de Biologia Molecular, Programa de Pós Graduação
em Engenharia de Bioprocessos e Biotecnologia, Universidade Federal do Paraná, Curitiba, Paraná 81531-990, Brazil
| | - Aline Cristina dos Santos
- Laboratório
Provas Biológicas, Instituto de Tecnologia
do Paraná, Curitiba, Paraná 80035-060, Brazil
| | - Odir Antônio Dellagostin
- Programa
de Pós Graduação em Biotecnologia, Centro de
Desenvolvimento Tecnológico, Universidade
Federal de Pelotas, Pelotas, Rio Grande do Sul 96160-000, Brazil
| | - Carlos Ricardo Soccol
- Laboratório
de Biologia Molecular, Programa de Pós Graduação
em Engenharia de Bioprocessos e Biotecnologia, Universidade Federal do Paraná, Curitiba, Paraná 81531-990, Brazil
| | - Vanete Thomaz-Soccol
- Laboratório
de Biologia Molecular, Programa de Pós Graduação
em Engenharia de Bioprocessos e Biotecnologia, Universidade Federal do Paraná, Curitiba, Paraná 81531-990, Brazil
| |
Collapse
|
8
|
Stari L, Jittayasotorn T, Inoue C, Chien MF. Complete genome sequence of a carbon tetrachloride-degrading bacterium, Pseudomonas sp. strain Stari2. Microbiol Resour Announc 2025; 14:e0095824. [PMID: 39878464 PMCID: PMC11895450 DOI: 10.1128/mra.00958-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/11/2025] [Indexed: 01/31/2025] Open
Abstract
This article reports on the complete genome of Pseudomonas sp. strain Stari2, which was shown to have the ability to degrade carbon tetrachloride (CCl4) in aerobic conditions. A single circular sequence of 6,310,573 bp, a GC content of 60.3%, and 5,680 coding sequences were obtained. In silico analysis revealed an average nucleotide identity of 94.06% to its closest species.
Collapse
Affiliation(s)
- Leonardo Stari
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
- Graduate School of Life Science, Tohoku University, Sendai, Japan
| | | | - Chihiro Inoue
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
| | - Mei-Fang Chien
- Graduate School of Environmental Studies, Tohoku University, Sendai, Japan
| |
Collapse
|
9
|
Dunkley ORS, Bell AG, Modi NH, Huang Y, Tseng S, Reiss R, Daivaa N, Davis JL, Vargas DA, Banada P, Xie YL, Myhrvold C. A Streamlined Point-of-Care CRISPR Test for Tuberculosis Detection Directly from Sputum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322517. [PMID: 40034782 PMCID: PMC11875272 DOI: 10.1101/2025.02.19.25322517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Mycobacterium tuberculosis (Mtb) is a major threat to global health and is responsible for over one million deaths each year. To stem the tide of cases and maximize opportunities for early interventions, there is an urgent need for affordable and simple means of tuberculosis diagnosis in under-resourced areas. We sought to develop a CRISPR-based isothermal assay coupled with a compatible, straightforward sample processing technique for point-of-care use. Here, we combine Recombinase Polymerase Amplification (RPA) with Cas13a and Cas12a, to create two parallelised one-pot assays that detect two conserved elements of Mtb (IS6110 and IS1081) and an internal control targeting human DNA. These assays were shown to be compatible with lateral flow and can be readily lyophilized. Our finalized assay exhibited sensitivity over a wide range of bacterial loads (105 to 102 CFU/mL) in sputum. The limit of detection (LoD) of the assay was determined to be 69.0 (51.0 - 86.9) CFU/mL for Mtb strain H37Rv spiked in sputum and 80.5 (59.4 - 101.6) CFU/mL for M. bovis BCG. Our assay showed no cross reactivity against a wide range of bacterial/fungal isolates. Clinical tests on 13 blinded sputum samples revealed 100% (6/6) sensitivity and 100% (7/7) specificity compared to culture. Our assay exhibited comparable sensitivity in clinical samples to the microbiological gold standard, TB culture, and to the nucleic acid state-of-the-art, GeneXpert MTB/RIF Ultra. This technology streamlines TB diagnosis from sample extraction to assay readout in a rapid and robust format, making it the first test to combine amplification and detection while being compatible with both lateral flow and lyophilization.
Collapse
Affiliation(s)
- Owen R S Dunkley
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Alexandra G Bell
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Nisha H Modi
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yujia Huang
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Soleil Tseng
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
| | - Robert Reiss
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Naranjargal Daivaa
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - J Lucian Davis
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Pulmonary, Critical Care, and Sleep Medicine Section, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Deninson Alejandro Vargas
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Padmapriya Banada
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Yingda L Xie
- Public Health Research Institute, Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Cameron Myhrvold
- Department of Molecular Biology, Princeton University, Princeton New Jersey, 08544, USA
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, New Jersey, 08544, USA
- Department of Chemistry, Princeton University, Princeton, New Jersey, 08544, USA
| |
Collapse
|
10
|
Santoshi M, Tare P, Nagaraja V. Nucleoid-associated proteins of mycobacteria come with a distinctive flavor. Mol Microbiol 2025; 123:177-194. [PMID: 38922783 DOI: 10.1111/mmi.15287] [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: 02/16/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
In every bacterium, nucleoid-associated proteins (NAPs) play crucial roles in chromosome organization, replication, repair, gene expression, and other DNA transactions. Their central role in controlling the chromatin dynamics and transcription has been well-appreciated in several well-studied organisms. Here, we review the diversity, distribution, structure, and function of NAPs from the genus Mycobacterium. We highlight the progress made in our understanding of the effects of these proteins on various processes and in responding to environmental stimuli and stress of mycobacteria in their free-living as well as during distinctive intracellular lifestyles. We project them as potential drug targets and discuss future studies to bridge the information gap with NAPs from well-studied systems.
Collapse
Affiliation(s)
- Meghna Santoshi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Priyanka Tare
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Valakunja Nagaraja
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| |
Collapse
|
11
|
Ashokcoomar S, Pillay M. Differential expression of genes associated with lipid import, β-oxidation and lactate oxidation induced by Mycobacterium tuberculosis curli pili in broth culture compared to intracellular bacilli within THP-1 macrophages. J Med Microbiol 2025; 74. [PMID: 40162564 DOI: 10.1099/jmm.0.001994] [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: 04/02/2025] Open
Abstract
Introduction. The adhesin, Mycobacterium tuberculosis curli pili (MTP), assists the pathogen in attachment, invasion and disease progression. Previously, this adhesin was demonstrated to contribute to the pathogen's cell wall functions and fatty acid metabolism and affects total metabolite abundance in central carbon metabolism and fatty acid metabolism of the host. The accumulation/depletion of metabolites is reliant on the gene expression of proteins involved in the import, transport and breakdown of substrates.Gap statement. MTP has not been investigated in relation to genes involved in import/transport/breakdown of substrates.Aim. This study aimed to investigate the possible regulatory role of MTP in modulating metabolic changes of the pathogen in different microenvironments.Methods. Ribonucleic acid was harvested from bacterial broth cultures of adhesin-proficient and adhesin-deficient M. tuberculosis. These strains were also used to infect differentiated THP-1 macrophages for 4 h prior to isolation of intracellular bacteria, RNA extraction and reverse transcription real-time quantitative PCR. The expression levels of selected genes involved in fatty acid transport (lucA, mce1D, mceG, Rv2799, Rv0966c and omamB), β-oxidation (fadA5 and fadB), lactate oxidation (lldD1 and lldD2) and gluconeogenic carbon flow (pckA) were analysed by absolute quantification.Results. The gene expression levels of lucA, mce1D and pckA were significantly lower, and those of Rv2799, Rv0966c, mceG, fadA5 and lldD2 were significantly higher in the adhesin-proficient cultured bacterial strains relative to the Δmtp strain. The intracellular adhesin-proficient bacteria displayed significantly higher gene expression levels of Rv2799 and significantly lower gene expression levels of Rv0966c, fadA5, lldD1 and pckA relative to the Δmtp strain. Interestingly, during early infection, the intracellular Δmtp displayed significantly increased expression of omamB, mceG, fadB, lldD1 and lldD2 relative to the broth culture. This trend was inverted in the WT models.Conclusion. MTP are significantly associated with the regulation of genes involved in lipid transport, β-oxidation and lactate oxidation.
Collapse
Affiliation(s)
- Shinese Ashokcoomar
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa
| | - Manormoney Pillay
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, 1st floor Doris Duke Medical Research Institute, Congella, Private Bag 7, Durban 4013, South Africa
| |
Collapse
|
12
|
Nelson DJ, Chugh K, Pua HH, Haselton FR. A Software Tool for Reagent Design to Expand Access to Single-Nucleotide Variant Detection by the Oligonucleotide Ligation Assay. J Mol Diagn 2025; 27:184-198. [PMID: 39818318 DOI: 10.1016/j.jmoldx.2024.12.007] [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: 05/21/2024] [Revised: 09/26/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025] Open
Abstract
Single-nucleotide variants (SNVs) and polymorphisms are characteristic biomarkers in various biological contexts, including pathogen drug resistances and human diseases. Tools that lower the implementation barrier of molecular SNV detection methods would provide greater leverage of the expanding single-nucleotide polymorphism/SNV database. The oligonucleotide ligation assay (OLA) is a highly specific means for detection of known SNVs and is especially powerful when coupled with PCR. Yet, the OLA design process remains intensive, and criteria for success are uncertain. To assist in the design process, this study describes OLAgen, an open-source tool to automate development of OLAs and their coupled PCR assays. The software facilitates alignment of sequences surrounding SNVs and generates ligation probes while screening for dimerization potential. OLAgen successfully produced ligation probes that closely matched previously validated designs for HIV-1, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and KRAS, confirming its reliability and potential for clinical applications. The tool was used to generate new assays targeting Mycobacterium tuberculosis drug resistance and variants in the human JAK2, BRAF, and factor V genes, all of which demonstrated 100% sensitivity and specificity in controlled laboratory experiments. The OLAgen predicted assay designs detected mutant frequencies as low as 1% to 5% in wild-type backgrounds in proof-of-concept laboratory studies. OLAgen represents a significant advancement in accessible assay design, promoting the broader application of OLA technology in clinical and research settings.
Collapse
Affiliation(s)
- Dalton J Nelson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kunal Chugh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Heather H Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Frederick R Haselton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
13
|
Chen B, Bajramović B, Vriesendorp B, Spaink HP. Evolution of the PE_PGRS Proteins of Mycobacteria: Are All Equal or Are Some More Equal than Others? BIOLOGY 2025; 14:247. [PMID: 40136504 PMCID: PMC11939664 DOI: 10.3390/biology14030247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
PE_PGRS domain proteins represent a family of proteins found in pathogenic and non-pathogenic mycobacteria such as M. smegmatis. This conserved family is characterized by two distinct regions denoted as the variable PGRS domain defined by glycine-rich repeats, and a PE domain consisting of two antiparallel alpha-helices. There are many indications that PE_PGRS proteins are involved in immunopathogenesis and virulence by evading or triggering the host immune response. However, there is not yet any information on their degree of specialization or redundancy. Computational analysis and structural annotation using AlphaFold3 combined with other tools reveals an exceptionally powerful and unprecedented ability to undergo phase separation by the PGRS domain. This suggests that PGRS's glycine-rich, multivalent, low-complexity composition supports phase separation while adopting a structured conformation, contrary to the disordered nature typical of such domains. While previously never reported, the hypothesized role of PGRS in virulence indicates a novel window into the seemingly ubiquitous role of phase separation in cellular compartmentalization and molecular dynamics. This review aims to summarize the current understanding of the PE_PGRS family and its various biological roles in the context of bioinformatic analyses of some interesting representatives of M. marinum that are under control by host sterols. Based on the structural bioinformatics analysis, we discuss future approaches to uncover the mechanistic role of this intriguing family of mycobacterial proteins in both pathogenic and non-pathogenic mycobacteria.
Collapse
Affiliation(s)
| | | | | | - Herman Pieter Spaink
- Institute of Biology, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (B.C.); (B.B.); (B.V.)
| |
Collapse
|
14
|
Xie W, Luo D, Soni V, Wang Z. Functional characterization of MMAR_1296 in Mycobacterium marinum and its potential as a vaccine candidate. Vaccine 2025; 48:126720. [PMID: 39809090 DOI: 10.1016/j.vaccine.2025.126720] [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: 10/08/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
The Pro-Glu/Pro-Pro-Glu (PE/PPE) family proteins in mycobacteria plays a crucial role in pathogenesis and immune evasion. These proteins characterized by unique structures with conserved sequences. This study elucidated the specific immunological functions of MMAR_1296 from marine mycobacterium. Expressing MMAR_1296 in Mycobacterium smegmatis (M. smegmatis) led to significant alterations in bacterial morphology, as well as reduced survival of M. smegmatis under adverse in vitro conditions and within macrophages. Furthermore, transcriptome analysis of mouse macrophages indicated that natural immunity-related pathways were upregulated in the group infected with M. smegmatis recombinantly expressing MMAR_1296. Moreover, the mycobacterium Growth Inhibition Assays(MGIA)in mice demonstrated that M. smegmatis expressing MMAR_1296 exerted a significant inhibitory effect against Mycobacterium abscessus (M. abscessus) and Mycobacterium marinum (M. marinum) infections. Immunization challenge experiments in mice further confirmed its protective effects, showing a reduction in organ bacterial loads by 1 log10 value compared to the positive control group. These findings indicate that MMAR_1296 is a promising vaccine candidate for M. abscessus and M. marinum. Given that PE/PPE protein family is also a crucial component of Mycobacterium tuberculosis (M. tuberculosis) antigens, further exploration of sequence functions based on MMAR_1296 could reveal broader applications of PE/PPE proteins family for M. tuberculosis treatment. This study supported vaccine development targeting PE/PPE proteins in mycobacteria and paves the way for broader applications.
Collapse
Affiliation(s)
- Weile Xie
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
15
|
Krysenko S, Emani CS, Bäuerle M, Oswald M, Kulik A, Meyners C, Hillemann D, Merker M, Prosser G, Wohlers I, Hausch F, Brötz-Oesterhelt H, Mitulski A, Reiling N, Wohlleben W. GlnA3 Mt is able to glutamylate spermine but it is not essential for the detoxification of spermine in Mycobacterium tuberculosis. J Bacteriol 2025; 207:e0043924. [PMID: 39882905 PMCID: PMC11841054 DOI: 10.1128/jb.00439-24] [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: 10/11/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
Mycobacterium tuberculosis is well adapted to survive and persist in the infected host, escaping the host's immune response. Since polyamines such as spermine, which are synthesized by infected macrophages, are able to inhibit the growth of M. tuberculosis, the pathogen needs strategies to cope with these toxic metabolites. The actinomycete Streptomyces coelicolor, a close relative of M. tuberculosis, makes use of a gamma-glutamylation pathway to functionally neutralize spermine. We therefore considered whether a similar pathway would be functional in M. tuberculosis. In the current study, we demonstrated that M. tuberculosis growth was inhibited by the polyamine spermine. Using in vitro enzymatic assays we determined that GlnA3Mt (Rv1878) possesses genuine gamma-glutamylspermine synthetase catalytic activity. We further showed that purified His-Strep-GlnA3Mt, as well as native GlnA3Mt, prefer spermine as a substrate over putrescine, cadaverine, spermidine, or other monoamines and amino acids, suggesting that GlnA3Mt may play a specific role in the detoxification of the polyamine spermine. However, the deletion of the glnA3 gene in M. tuberculosis did not result in growth inhibition or enhanced sensitivity of M. tuberculosis in the presence of high spermine concentrations. Gene expression analysis of spermine-treated M. tuberculosis revealed no difference in the level of glnA3Mt expression relative to untreated cells, whereas a gene encoding a previously characterized efflux pump (Mmr; rv3065) was significantly upregulated. This suggests that bacterial survival under elevated spermine concentrations can not only be achieved by detoxification of spermine itself but also by mechanisms resulting in decreased spermine levels in the bacteria. IMPORTANCE Upon Mycobacterium tuberculosis infection macrophages synthesize the polyamine spermine, which at elevated concentrations is toxic for M. tuberculosis. Based on our investigations of spermine resistance in the closely related actinomycete Streptomyces coelicolor, we hypothesized that the glutamylspermine synthetase GlnA3 may be responsible for the resistance of M. tuberculosis against toxic spermine. Here we show that GlnA3Mt can indeed covalently modify spermine via glutamylation. However, GlnA3Mt is probably not the only resistance mechanism since a glnA3 null mutant of M. tuberculosis can survive under spermine stress. Gene expression studies suggest that an efflux pump may participate in resistance. Thus a combination of GlnA3Mt and specific efflux pumps acting as putative spermine transporters may constitute an active spermine-detoxification system in M. tuberculosis.
Collapse
Affiliation(s)
- Sergii Krysenko
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Carine Sao Emani
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
| | - Moritz Bäuerle
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Maria Oswald
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Andreas Kulik
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Christian Meyners
- Institute of Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Hessen, Germany
| | - Doris Hillemann
- National Reference Center for Mycobacteria, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
| | - Matthias Merker
- Evolution of the Resistome, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
| | - Gareth Prosser
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
| | - Inken Wohlers
- Data Science, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
| | - Felix Hausch
- Institute of Organic Chemistry and Biochemistry, Technical University Darmstadt, Darmstadt, Hessen, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Hessen, Germany
| | - Heike Brötz-Oesterhelt
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbial Bioactive Compounds, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Agnieszka Mitulski
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbial Bioactive Compounds, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Norbert Reiling
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Schleswig-Holstein, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | - Wolfgang Wohlleben
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), Department of Microbiology and Biotechnology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Baden-Württemberg, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| |
Collapse
|
16
|
Ramesh M, Behra PRK, Pettersson BMF, Dasgupta S, Kirsebom LA. Age-Dependent Pleomorphism in Mycobacterium monacense Cultures. Microorganisms 2025; 13:475. [PMID: 40142368 PMCID: PMC11946739 DOI: 10.3390/microorganisms13030475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Changes in cell shape have been shown to be an integral part of the mycobacterial life cycle; however, systematic investigations into its patterns of pleomorphic behaviour in connection with stages or conditions of growth are scarce. We have studied the complete growth cycle of Mycobacterium monacense cultures, a Non-Tuberculous Mycobacterium (NTM), in solid as well as in liquid media. We provide data showing changes in cell shape from rod to coccoid and occurrence of refractive cells ranging from Phase Grey to phase Bright (PGB) in appearance upon ageing. Changes in cell shape could be correlated to the bi-phasic nature of the growth curves for M. monacense (and the NTM Mycobacterium boenickei) as measured by the absorbance of liquid cultures while growth measured by colony-forming units (CFU) on solid media showed a uniform exponential growth. Based on the complete M. monacense genome we identified genes involved in cell morphology, and analyses of their mRNA levels revealed changes at different stages of growth. One gene, dnaK_3 (encoding a chaperone), showed significantly increased transcript levels in stationary phase cells relative to exponentially growing cells. Based on protein domain architecture, we identified that the DnaK_3 N-terminus domain is an MreB-like homolog. Endogenous overexpression of M. monacense dnaK_3 in M. monacense was unsuccessful (appears to be lethal) while exogenous overexpression in Mycobacterium marinum resulted in morphological changes with an impact on the frequency of appearance of PGB cells. However, the introduction of an anti-sense "gene" targeting the M. marinum dnaK_3 did not show significant effects. Using dnaK_3-lacZ reporter constructs we also provide data suggesting that the morphological differences could be due to differences in the regulation of dnaK_3 in the two species. Together these data suggest that, although its regulation may vary between mycobacterial species, the dnaK_3 might have a direct or indirect role in the processes influencing mycobacterial cell shape.
Collapse
Affiliation(s)
| | | | | | | | - Leif A. Kirsebom
- Department of Cell and Molecular Biology, Biomedical Centre, Box 596, SE-751 24 Uppsala, Sweden; (M.R.); (P.R.K.B.); (B.M.F.P.); (S.D.)
| |
Collapse
|
17
|
Dokas S, Taylor DK, Good LL, Mohanaraj S, Maillard RA. Identifying Allosteric Hotspots in Mycobacterium tuberculosis cAMP Receptor Protein through Structural Homology. Biochemistry 2025; 64:801-811. [PMID: 39887300 PMCID: PMC11840924 DOI: 10.1021/acs.biochem.4c00723] [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: 10/27/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Understanding the mechanisms of allosteric regulation in response to second messengers is crucial for advancing basic and applied research. This study focuses on the differential allosteric regulation by the ubiquitous signaling molecule, cAMP, in the cAMP receptor protein from Escherichia coli (CRPEcoli) and from Mycobacterium tuberculosis (CRPMTB). By introducing structurally homologous mutations from allosteric hotspots previously identified in CRPEcoli into CRPMTB and examining their effects on protein solution structure, stability and function, we aimed to determine the factors contributing to their differential allosteric regulation. Our results demonstrate that the mutations did not significantly alter the overall fold, assembly and thermodynamic stability of CRPMTB, but had varying effects on cAMP binding affinity and cooperativity. Interestingly, the mutations had minimal impact on the specific binding of CRPMTB to DNA promoter sites. However, we found that cAMP primarily reduces nonspecific CRPMTB-DNA complexes and that the mutants largely lose this ability. Furthermore, our experiments revealed that CRPMTB-DNA complexes serve as a nucleation point for additional binding of CRPMTB proteins to form high-order oligomers with the DNA. Overall, our findings highlight the importance of both cAMP and DNA interactions in modulating the allosteric regulation of CRPMTB and provide insights into the differential responses of CRPEcoli and CRPMTB to cAMP.
Collapse
Affiliation(s)
- Stephen
P. Dokas
- Department
of Chemistry, Georgetown University, Washington, District of
Columbia 20057, United States
| | - Daniel K. Taylor
- Department
of Chemistry, Georgetown University, Washington, District of
Columbia 20057, United States
| | - Lydia L. Good
- Department
of Chemistry, Georgetown University, Washington, District of
Columbia 20057, United States
| | - Sanuja Mohanaraj
- Department
of Chemistry, Georgetown University, Washington, District of
Columbia 20057, United States
| | - Rodrigo A. Maillard
- Department
of Chemistry, Georgetown University, Washington, District of
Columbia 20057, United States
- Institute
of Soft Matter Synthesis and Metrology, Georgetown University, Washington, District of Columbia 20057, United States
| |
Collapse
|
18
|
Rai R, Paroha R, Rai S, Singh AK, Chaurasia R, Agarwal N, Pandey MK, Chaurasiya SK. Cloning, expression, purification, and characterization of glutamate decarboxylase (Rv3432c) from Mycobacterium tuberculosis. Int Microbiol 2025:10.1007/s10123-025-00637-8. [PMID: 39954146 DOI: 10.1007/s10123-025-00637-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
Glutamate decarboxylase (Gad), a pyridoxal 5'-phosphate (PLP)-dependent enzyme, catalyzes the conversion of glutamate to γ-aminobutyric acid (GABA), consuming a proton in the process and thereby contributing to intracellular pH homeostasis in bacteria. However, the presence and function of the Gad-dependent mechanism in mycobacteria remain largely unexplored. This study aimed to characterize Gad activity in Mycobacterium tuberculosis (Mtb). We detected Gad activity in live cells of both Mtb and Mycobacterium smegmatis (MS). Gad activity and GABA was also detected in cell lysates of Mtb and MS. The gadB gene from Mtb was cloned, expressed, and GadB protein was purified under native conditions using MS as an expression host. Initial attempts to express GadB in Escherichia coli (E. coli) resulted in the overexpressed protein being present in the insoluble fraction and was enzymatically inactive when purified under denaturing conditions. Subsequently, an acetamide-inducible expression system was employed in MS for successful overexpression and purification of recombinant GadB. 6 × His-GadB was purified using immobilized metal affinity chromatography, and its molecular weight was determined to be ~ 51.2 kDa by SDS-PAGE. The purified 6 × His-GadB enzyme was active at both neutral and acidic pH. Its activity was found to be PLP-dependent, with optimal activity at pH 7.2 and 50°C. These findings suggest that Gad is expressed in Mtb both in normal and in acidic medium, supporting the possible existence of a Gad-dependent acid resistance mechanism in Mtb.
Collapse
Affiliation(s)
- Rupal Rai
- Molecular Signalling Lab, Department of Biological Sciences and Engineering, Maulana Azad National Institute of Technology Bhopal, Bhopal, M.P, India
| | - Ruchi Paroha
- Microbiology and Immunology Department, UTMB, Galveston, TX, USA
| | | | - Anirudh K Singh
- School of Sciences, SAM Global University, Raisen, M.P, India
| | - Rashmi Chaurasia
- Department of Basic Science, IES College of Technology, Bhopal, M.P, India
| | | | - Megha Katare Pandey
- Translational Medicine, All India Institute of Medical Sciences, Bhopal, M.P, India
| | - Shivendra K Chaurasiya
- Molecular Signalling Lab, Department of Biological Sciences and Engineering, Maulana Azad National Institute of Technology Bhopal, Bhopal, M.P, India.
| |
Collapse
|
19
|
Mitton-Fry MJ, Cummings JE, Lu Y, Armenia JF, Byl JAW, Oviatt AA, Bauman AA, Robertson GT, Osheroff N, Slayden RA. Anti-Mycobacterial Activity of Bacterial Topoisomerase Inhibitors with Dioxygenated Linkers. ACS Infect Dis 2025; 11:474-482. [PMID: 39792935 PMCID: PMC11834967 DOI: 10.1021/acsinfecdis.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Developing new classes of drugs that are active against infections caused by Mycobacterium tuberculosis is a priority for treating and managing this deadly disease. Here, we describe screening a small library of 20 DNA gyrase inhibitors and identifying new lead compounds. Three structurally diverse analogues were identified with minimal inhibitory concentrations of 0.125 μg/mL against both drug-susceptible and drug-resistant strains of M. tuberculosis. These lead compounds also demonstrated antitubercular activity in ex vivo studies using infected THP-1 macrophages with minimal cytotoxicity in THP-1, HeLa, and HepG2 cells (IC50 ≥ 128 μg/mL). The molecular target of the lead compounds was validated through biochemical studies of select analogues with purified M. tuberculosis gyrase and the generation of resistant mutants. The lead compounds were assessed in combination with bedaquiline and pretomanid to determine the clinical potential, and the select lead (158) demonstrated in vivo efficacy in an acute model of TB infection in mice, reducing the lung bacterial burden by approximately 3 log10 versus untreated control mice. The advancement of DNA gyrase inhibitors expands the field of innovative therapies for tuberculosis and may offer an alternative to fluoroquinolones in future therapeutic regimens.
Collapse
Affiliation(s)
- Mark J. Mitton-Fry
- Division
of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jason E. Cummings
- Department
of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Yanran Lu
- Division
of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jillian F. Armenia
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jo Ann W. Byl
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Alexandria A. Oviatt
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Allison A. Bauman
- Department
of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Gregory T. Robertson
- Department
of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Neil Osheroff
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Medicine (Hematology/Oncology), Vanderbilt
University School of Medicine, Nashville, Tennessee 37232, United States
| | - Richard A. Slayden
- Department
of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523, United States
| |
Collapse
|
20
|
Lynde B, Chemaly DM, Baldin VP, Greve E, Harding CL, Graner JM, Hardy M, Chowdhury S, Parish T. Novel 3-Aminothieno[2,3- b]pyridine-2-carboxamides with Activity against Mycobacterium tuberculosis. ACS Med Chem Lett 2025; 16:241-249. [PMID: 39967629 PMCID: PMC11831384 DOI: 10.1021/acsmedchemlett.4c00472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025] Open
Abstract
We conducted an exploration of the 3-aminothieno[2,3-b]pyridine-2-carboxamide (TPA) series for its potential as a drug scaffold against Mycobacterium tuberculosis. Existing analogs were active against a recombinant strain of M. tuberculosis with reduced expression of the sole signal peptidase LepB, but with poor activity against the wild-type strain. Our aim was to improve potency and explore the structure-activity relationship of the series. We identified two subsets of TPA. The first subset of compounds had equipotent activity against wild-type and LepB hypomorph strains and may represent a series with a different target. The second subset of compounds had increased activity against the LepB hypomorph strain and thus appears to be pathway-specific. Among this latter set we identified 17af as a potent inhibitor (IC90 = 1.2 μM) with some cytotoxicity (IC50 = 19 μM) and which retained increased activity against the LepB hypomorph (IC90 = 0.41 μM).
Collapse
Affiliation(s)
- Brock
E. Lynde
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Danielle M. Chemaly
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Vanessa Pietrowski Baldin
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Eric Greve
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Christopher L. Harding
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Jasmin M. Graner
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Mason Hardy
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Sultan Chowdhury
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
| | - Tanya Parish
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98109, United States
- Department
of Pediatrics, University of Washington
School of Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
21
|
Wynn EA, Dide-Agossou C, Al Mubarak R, Rossmassler K, Ektnitphong V, Bauman AA, Massoudi LM, Voskuil MI, Robertson GT, Moore CM, Walter ND. Emergence of antibiotic-specific Mycobacterium tuberculosis phenotypes during prolonged treatment of mice. Antimicrob Agents Chemother 2025; 69:e0131024. [PMID: 39818957 PMCID: PMC11823617 DOI: 10.1128/aac.01310-24] [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: 09/04/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025] Open
Abstract
A major challenge in tuberculosis (TB) therapeutics is that antibiotic exposure leads to changes in the physiology of M. tuberculosis (Mtb), which may enable the pathogen to withstand treatment. While antibiotic-treated Mtb has been evaluated in in vitro experiments, it is unclear if and how long-term in vivo treatment with diverse antibiotics with varying treatment-shortening activity (sterilizing activity) affects Mtb physiologic processes differently. Here, we used SEARCH-TB, a pathogen-targeted RNA-sequencing platform, to characterize the Mtb transcriptome in the BALB/c high-dose aerosol infection mouse model following 4 weeks of treatment with three sterilizing and three non-sterilizing antibiotics. Certain transcriptional changes were shared among most antibiotics, including decreased expression of genes associated with protein synthesis and metabolism and the induction of certain genes associated with stress responses. However, the magnitude of this shared response differed between antibiotics. Sterilizing antibiotics rifampin, pyrazinamide, and bedaquiline generated a more quiescent Mtb state than did non-sterilizing antibiotics isoniazid, ethambutol, and streptomycin, as indicated by the decreased expression of genes associated with translation, transcription, secretion of immunogenic proteins, metabolism, and cell wall synthesis. Additionally, we identified distinguishing transcriptional effects specific to each antibiotic, indicating that different mechanisms of action induce distinct patterns in response to cellular injury. In addition to elucidating the Mtb physiologic changes associated with antibiotic stress, this study demonstrates the value of SEARCH-TB as a highly granular pharmacodynamic assay that reveals antibiotic effects that are not apparent based on culture alone.
Collapse
Affiliation(s)
- Elizabeth A. Wynn
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
| | - Christian Dide-Agossou
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Reem Al Mubarak
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karen Rossmassler
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Victoria Ektnitphong
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Allison A. Bauman
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Lisa M. Massoudi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Martin I. Voskuil
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Gregory T. Robertson
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Camille M. Moore
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nicholas D. Walter
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
22
|
Dixit A, Ektefaie Y, Kagal A, Freschi L, Karyakarte R, Lokhande R, Groschel M, Tornheim JA, Gupte N, Pradhan NN, Paradkar MS, Deshmukh S, Kadam D, Schito M, Engelthaler DM, Gupta A, Golub J, Mave V, Farhat M. Drug Resistance and Epidemiological Success of Modern Mycobacterium tuberculosis Lineages in Western India. J Infect Dis 2025; 231:84-93. [PMID: 38819323 PMCID: PMC11793027 DOI: 10.1093/infdis/jiae240] [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: 08/11/2023] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Drivers of tuberculosis (TB) transmission in India, the country estimated to carry a quarter of the world's burden, are not well studied. We conducted a genomic epidemiology study to compare epidemiological success, host factors, and drug resistance among the 4 major Mycobacterium tuberculosis (Mtb) lineages (L1-L4) circulating in Pune, India. METHODS We performed whole-genome sequencing (WGS) of Mtb sputum culture-positive isolates from participants in two prospective cohort studies and predicted genotypic susceptibility using a validated random forest model. We compared lineage-specific phylogenetic and time-scaled metrics to assess epidemiological success. RESULTS Of the 612 isolates that met sequence quality criteria, Most were L3 (44.6%). The majority (61.1%) of multidrug-resistant isolates were L2 (P < .001) and L2 demonstrated a higher rate and more recent resistance acquisition. L4 and/or L2 demonstrated higher clustering and time-scaled haplotypic density (THD) compared to L3 and/or L1, suggesting higher epidemiological success. L4 demonstrated higher THD and clustering (odds ratio, 5.1 [95% confidence interval, 2.3-12.3]) in multivariate models controlling for host factors and resistance. CONCLUSIONS L2 shows a higher frequency of resistance, and both L2 and L4 demonstrate evidence of higher epidemiological success than L3 or L1 in Pune. Contact tracing around TB cases and heightened surveillance of TB DR in India is a public health priority.
Collapse
Affiliation(s)
- Avika Dixit
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasha Ektefaie
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Anju Kagal
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Luca Freschi
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Rahul Lokhande
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | - Matthias Groschel
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey A Tornheim
- Center for Clinical Global Health Education, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nikhil Gupte
- Center for Clinical Global Health Education, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Byramjee-Jeejeebhoy Medical College–Johns Hopkins University Clinical Trials Unit, Pune, India
- Johns Hopkins India, Pune, India
| | - Neeta N Pradhan
- Byramjee-Jeejeebhoy Medical College–Johns Hopkins University Clinical Trials Unit, Pune, India
- Johns Hopkins India, Pune, India
| | - Mandar S Paradkar
- Byramjee-Jeejeebhoy Medical College–Johns Hopkins University Clinical Trials Unit, Pune, India
- Johns Hopkins India, Pune, India
| | - Sona Deshmukh
- Byramjee-Jeejeebhoy Medical College–Johns Hopkins University Clinical Trials Unit, Pune, India
- Johns Hopkins India, Pune, India
| | - Dileep Kadam
- Byramjee-Jeejeebhoy Government Medical College, Pune, India
| | | | | | - Amita Gupta
- Center for Clinical Global Health Education, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jonathan Golub
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vidya Mave
- Center for Clinical Global Health Education, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Byramjee-Jeejeebhoy Medical College–Johns Hopkins University Clinical Trials Unit, Pune, India
- Johns Hopkins India, Pune, India
| | - Maha Farhat
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Davis ET, Raman R, Byrne SR, Ghanegolmohammadi F, Mathur C, Begley U, Dedon PC, Begley TJ. Genes and Pathways Comprising the Human and Mouse ORFeomes Display Distinct Codon Bias Signatures that Can Regulate Protein Levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636209. [PMID: 39974974 PMCID: PMC11838421 DOI: 10.1101/2025.02.03.636209] [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: 02/21/2025]
Abstract
Arginine, glutamic acid and selenocysteine based codon bias has been shown to regulate the translation of specific mRNAs for proteins that participate in stress responses, cell cycle and transcriptional regulation. Defining codon-bias in gene networks has the potential to identify other pathways under translational control. Here we have used computational methods to analyze the ORFeome of all unique human (19,711) and mouse (22,138) open-reading frames (ORFs) to characterize codon-usage and codon-bias in genes and biological processes. We show that ORFeome-wide clustering of gene-specific codon frequency data can be used to identify ontology-enriched biological processes and gene networks, with developmental and immunological programs well represented for both humans and mice. We developed codon over-use ontology mapping and hierarchical clustering to identify multi-codon bias signatures in human and mouse genes linked to signaling, development, mitochondria and metabolism, among others. The most distinct multi-codon bias signatures were identified in human genes linked to skin development and RNA metabolism, and in mouse genes linked to olfactory transduction and ribosome, highlighting species-specific pathways potentially regulated by translation. Extreme codon bias was identified in genes that included transcription factors and histone variants. We show that re-engineering extreme usage of C- or U-ending codons for aspartic acid, asparagine, histidine and tyrosine in the transcription factors CEBPB and MIER1, respectively, significantly regulates protein levels. Our study highlights that multi-codon bias signatures can be linked to specific biological pathways and that extreme codon bias with regulatory potential exists in transcription factors for immune response and development.
Collapse
Affiliation(s)
| | - Rahul Raman
- The RNA Institute, University at Albany, Albany, NY
- Department of Biological Sciences, University at Albany, Albany, NY
- Department of Biological Engineering and Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Shane R. Byrne
- Department of Biological Engineering and Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Farzan Ghanegolmohammadi
- Department of Biological Engineering and Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Chetna Mathur
- The RNA Institute, University at Albany, Albany, NY
- Department of Biological Sciences, University at Albany, Albany, NY
| | - Ulrike Begley
- The RNA Institute, University at Albany, Albany, NY
- Department of Biological Sciences, University at Albany, Albany, NY
| | - Peter C. Dedon
- Department of Biological Engineering and Center for Environmental Health Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, 138602, Singapore
| | - Thomas J. Begley
- The RNA Institute, University at Albany, Albany, NY
- Department of Biological Sciences, University at Albany, Albany, NY
- RNA Epitranscriptomics and Proteomics Resource, University at Albany, Albany, NY
| |
Collapse
|
24
|
Bahl A, Rakshit R, Pandey S, Tripathi D. Genome wide screening to discover novel toxin-antitoxin modules in Mycobacterium indicus pranii; perspective on gene acquisition during mycobacterial evolution. Biotechnol Appl Biochem 2025; 72:116-137. [PMID: 39113212 DOI: 10.1002/bab.2651] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/24/2024] [Indexed: 02/06/2025]
Abstract
Mycobacterium indicus pranii (MIP), a benign saprophyte with potent immunomodulatory attributes, holds a pivotal position in mycobacterial evolution, potentially serving as the precursor to the pathogenic Mycobacterium avium complex (MAC). Despite its established immunotherapeutic efficacy against leprosy and notable outcomes in gram-negative sepsis and COVID-19 cases, the genomic and biochemical features of MIP remain largely elusive. This study explores the uncharted territory of toxin-antitoxin (TA) systems within MIP, hypothesizing their role in mycobacterial pathogenicity regulation. Genome-wide screening, employing diverse databases, unveils putative TA modules in MIP, setting the stage for a comparative analysis with known modules in Mycobacterium tuberculosis, Mycobacterium smegmatis, Escherichia coli, and Vibrio cholerae. The study further delves into the TA network of MAC and Mycobacterium intracellulare, unraveling interactive properties and family characteristics of identified TA modules in MIP. This comprehensive exploration seeks to illuminate the contribution of TA modules in regulating virulence, habitat diversification, and the evolutionary pathogenicity of mycobacteria. The insights garnered from this investigation not only enhance our understanding of MIP's potential as a vaccine candidate but also hold promise in optimizing tuberculosis drug regimens for expedited recovery.
Collapse
Affiliation(s)
- Aayush Bahl
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Roopshali Rakshit
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Saurabh Pandey
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, Delhi, India
| | - Deeksha Tripathi
- Microbial Pathogenesis and Microbiome Lab, Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
25
|
Dahiya P, Bisht MK, Mukhopadhyay S. Role of PE family of proteins in mycobacterial virulence: Potential on anti-TB vaccine and drug design. Int Rev Immunol 2025:1-16. [PMID: 39889764 DOI: 10.1080/08830185.2025.2455161] [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: 07/23/2024] [Revised: 11/19/2024] [Accepted: 01/02/2025] [Indexed: 02/03/2025]
Abstract
Macrophages are the primary targets of mycobacterial infection, which plays crucial roles both in nonspecific defence (innate immunity) as well as specific defence mechanisms (adaptive immunity) by secreting various cytokines, antimicrobial mediators and presenting antigens to T-cells. Sequencing of the mycobacterial genome revealed that 10% of its coding ability is devoted to the Pro-Glu motif-containing (PE) and Pro-Pro-Glu motif-containing (PPE) family proteins. While the function of most of the genes belonging to the PE-PPE family initially remained unannotated, recent studies have shown that many proteins of this family play critical roles in bacterial growth and cell functions, and manipulation of host immune responses, indicating their potential roles in mycobacterial virulence. In this review, we have focussed on describing the immunological importance of particularly the PE group of proteins in the context of 'virulence' determinants and outcome of tuberculosis disease. Additionally, we have discussed about the roles of these proteins on host-pathogen-interaction and how some of these genes can be targeted which may help us in designing effective anti-TB therapeutics.
Collapse
Affiliation(s)
- Priyanka Dahiya
- Laboratory of Molecular Cell Biology, BRIC-Center for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
- Graduate Studies, Regional Center for Biotechnology, Faridabad, Haryana, India
| | - Manoj Kumar Bisht
- Laboratory of Molecular Cell Biology, BRIC-Center for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, BRIC-Center for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| |
Collapse
|
26
|
Gurmessa SK, Choi HG, Back YW, Jiang Z, Pham TA, Choi S, Kim HJ. Novel fusion protein REA induces robust prime protection against tuberculosis in mice. NPJ Vaccines 2025; 10:20. [PMID: 39890787 PMCID: PMC11785989 DOI: 10.1038/s41541-025-01077-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
While many novel candidates for tuberculosis vaccines are presently undergoing pre-clinical or clinical trials, none of them have been able to eliminate infection entirely. In this study, we engineered a potent chimeric protein vaccine candidate, Rv2299cD2D3-ESAT-6-Ag85B (REA), which induced Th1 and Th17 responses via dendritic cell maturation. REA-activated macrophages operated the killing mechanisms of Mycobacterium tuberculosis (MTB), such as phagosomal maturation and phagolysosome fusion, through the (PI3K)-p38 MAPK-Ca2+-NADPH oxidase pathway. Dendritic cells and macrophages activated by REA elicited synergistic anti-mycobacterial responses. Notably, REA-immunized mice suppressed MTB growth to undetectable levels at 16 weeks post-infection, which was supported by gross and pathologic findings and acid-fast staining of the lung tissues, and maintained antigen-specific multifunctional IFN-γ+IL-2+TNF-α CD4+ T and long-lasting T cells producing cytokines in the tissues. Our findings suggest that REA is an outstanding prime prophylactic vaccine candidate against tuberculosis.
Collapse
Affiliation(s)
- Sintayehu Kebede Gurmessa
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Han-Gyu Choi
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yong Woo Back
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Zongyou Jiang
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Thuy An Pham
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Seunga Choi
- R&D Center, Myco-Rapha Inc., Daejeon, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
27
|
Berida T, Huang TY, Weck SC, Lutz M, McKee SR, Kagerah N, Manning D, Jahan ME, Mishra SK, Doerksen RJ, Stallings CL, Ducho C, Roy S. 1,2,4-Triazole-based first-in-class non-nucleoside inhibitors of bacterial enzyme MraY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635793. [PMID: 39975250 PMCID: PMC11838528 DOI: 10.1101/2025.01.30.635793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
MraY, a bacterial enzyme crucial for the synthesis of peptidoglycans, represents a promising yet underexplored target for the development of effective antibacterial agents. Nature has provided several classes of nucleoside inhibitors of MraY and scientists have modified these structures further to obtain natural product-like inhibitors of MraY. The natural products and their synthetic analogs suffer from non-optimal in vivo efficacy, and the synthetic complexity of the structures renders the synthesis and structure-activity relationship (SAR) studies of these molecules particularly challenging. In this study, we present our findings on the discovery of first-in-class 1,2,4-triazole-based MraY inhibitors that are not nucleoside-derived. A series of 1,2,4-triazole analogous were identified by a structure-activity-relationship (SAR) study using a structure-based drug design strategy. Compound 1 , with an IC 50 of 171 µM against MraY from Staphylococcus aureus (MraY SA ), was optimized to compound 12a , exhibiting an IC 50 of 25 µM. Molecular docking studies against MraY SA provided insights into these compounds' binding interactions and activity. Furthermore, screening against the ESKAPE bacterial panel was also conducted, through which we discovered compounds demonstrating broad-spectrum antibacterial activity against E. faecium , methicillin-resistant S. aureus (MRSA), vancomycin-resistant Enterococci (VRE) strains and Mycobacterium tuberculosis . The novel, first-in-class non-nucleoside inhibitors of MraY highlighted in this work provide a strong proof-of-concept of how to leverage structural information of the protein to develop future antibacterial agents targeting MraY. Abstract Figure
Collapse
|
28
|
Jain M, Vyas R. Unveiling the silent defenders: mycobacterial stress sensors at the forefront to combat tuberculosis. Crit Rev Biotechnol 2025:1-19. [PMID: 39880585 DOI: 10.1080/07388551.2024.2449367] [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: 01/24/2024] [Revised: 07/12/2024] [Accepted: 09/14/2024] [Indexed: 01/31/2025]
Abstract
The global escalation in tuberculosis (TB) cases accompanied by the emergence of multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains of Mycobacterium tuberculosis (M.tb) emphasizes the critical requirement for novel potent drugs. The M.tb demonstrates extraordinary adaptability, thriving in diverse conditions, and always finds itself in win-win situations regardless of whether the environment is favorable or unfavorable; no matter the magnitude of the challenge, it can endure and survive. This review aims to uncover the role of multiple stress sensors of M.tb that assist bacteria in remaining viable within the host for years against various physiological stresses offered by the host. M.tb is an exceptionally triumphant pathogen, primarily due to its adeptness in developing defense mechanisms against stressful situations. The recent advances emphasize the significance of M.tb stress sensors, including chaperones, proteases, transcription factors, riboswitches, inteins, etc., employed in responding to a spectrum of physiological stresses imposed by the host, encompassing surface stress, host immune responses, osmotic stress, oxidative and nitrosative stresses, cell envelope stress, environmental stress, reductive stress, and drug pressure. These sensors act as silent defenders orchestrating adaptive strategies, with limited comprehensive information in current literature, necessitating a focused review. The M.tb strategies utilizing these stress sensors to mitigate the impact of traumatic conditions demand attention to neutralize this pathogen effectively. Moreover, the intricacies of these stress sensors provide potential targets to design an effective TB drug using structure-based drug design against this formidable global health threat.
Collapse
Affiliation(s)
- Manya Jain
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| | - Rajan Vyas
- Department of Life Sciences, Shiv Nadar Institution of Eminence (Deemed to be University), Gautam Buddha Nagar, Uttar Pradesh, India
| |
Collapse
|
29
|
Morey-León G, Fernández-Cadena JC, Andrade-Molina D, Berná L. Decoding Ecuadorian Mycobacterium tuberculosis Isolates: Unveiling Lineage-Associated Signatures in Beta-Lactamase Resistance via Pangenome Analysis. Biomedicines 2025; 13:313. [PMID: 40002726 PMCID: PMC11853040 DOI: 10.3390/biomedicines13020313] [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: 12/26/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Tuberculosis is the second largest public health threat caused by pathogens. Understanding Mycobacterium tuberculosis's transmission, virulence, and resistance profile is crucial for outbreak control. This study aimed to investigate the pangenome composition of Mycobacterium tuberculosis clinical isolates classified as L4 derived from Ecuador. Methods: We analyzed 88 clinical isolates of Mycobacterium tuberculosis by whole-genome sequencing (WGS) and bioinformatic tools for Lineage, Drug-resistance and Pangenome analysis. Results: In our analysis, we identified the dominance of the LAM lineage (44.3%). The pangenomic analysis revealed a core genome of approximately 3200 genes and a pangenome that differed in accessory and unique genes. According to the COG database, metabolism-related genes were the most representative of all partitions. However, differences were found within all lineages analyzed in the metabolic pathways described by KEGG. Isolates from Ecuador showed variations in genomic regions associated with beta-lactamase susceptibility, potentially leading to epistatic resistance to other drugs commonly used in TB treatment, warranting further investigation. Conclusions: Our findings provide valuable insights into the genetic diversity of Mycobacterium tuberculosis populations in Ecuador. These insights may be associated with increasing adaptation within host heterogeneity, variable latency periods, and reduced host damage, collectively contributing to disease spread. The application of WGS is essential to elucidating the epidemiology of TB in the country.
Collapse
Affiliation(s)
- Gabriel Morey-León
- Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
- Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
| | | | - Derly Andrade-Molina
- Facultad de Ciencias de la Salud, Universidad Espíritu Santo, Samborondón 0901952, Ecuador
- Laboratorio de Ciencias Ómicas, Universidad Espíritu Santo, Samborondón 0901952, Ecuador;
| | - Luisa Berná
- Laboratorio de Interacciones Hospedero-Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Unidad de Genómica Evolutiva, Facultad de Ciencias, Universidad de la República, Montevideo 11400, Uruguay
- Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| |
Collapse
|
30
|
Yun JS, Shin E, Lee YR, Lee JA, Lee H, Kim JS, Shin SJ, Ha SJ, Lee SW, Kim D, Yoo JS, Jeong HS. Immunogenicity and protective efficacy of a multi-antigenic adenovirus-based vaccine candidate against Mycobacterium tuberculosis. Front Microbiol 2025; 16:1492268. [PMID: 39927262 PMCID: PMC11802578 DOI: 10.3389/fmicb.2025.1492268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Introduction The inadequate efficacy of the Bacillus Calmette-Guérin (BCG) vaccine against adult pulmonary tuberculosis (TB) necessitates the development of new and effective vaccines. Human adenovirus serotype 5 (Ad5), which induces T-cell response, is a widely used viral vector. In this study, we aimed to evaluate the efficacy of a multi-antigenic recombinant Ad5 vectored vaccine and determine the optimal immunization route for enhanced immune response against Mycobacterium tuberculosis. Methods We constructed a multi-antigenic recombinant Ad5 vectored vaccine expressing four antigens (Ag85B-ESAT6-MPT64-Rv2660c) of M. tuberculosis (rAd-TB4), immunized with rAd-TB4 (5 × 107 infectious virus units/mouse) twice at an interval of 4 weeks starting at 10 weeks after BCG priming, and evaluated its boosting efficacy in a BCG-primed mouse model, and determined the optimal immunization route. Results Compared with the BCG-only (2 × 105 colony forming units/mouse), subcutaneous injection of rAd-TB4 (1 × 107 infectious virus units/mL; two doses) elicited a T-cell response and cytokine production in lung lymphocytes and splenocytes. rAd-TB4 immunization significantly reduced bacterial loads and inflamed lung areas compared to BCG immunization (p < 0.01) and protected against the H37Rv challenge performed at 17 weeks of BCG priming. RNA sequencing of the whole blood of rAd-TB4-vaccinated mice collected pre- and, 1 and 4 weeks post-infection, identified differentially expressed genes associated with immune and inflammatory responses, especially those in the Wnt signaling pathway. Conclusion Our results indicate that rAd-TB4 immunization enhances the immune response to the vaccine boosting antigens in BCG-primed mice, making it a potential adult pulmonary TB vaccine candidate.
Collapse
Affiliation(s)
- Jin-Seung Yun
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Eunkyung Shin
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Young-Ran Lee
- Bio-Convergence R&D Division, Korea Institute of Ceramic Engineering and Technology, Cheongju, Chungbuk, Republic of Korea
| | - Jung-Ah Lee
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hyeokjin Lee
- Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jong-Seok Kim
- Department of Cell Biology, College of Medicine, Myunggok Medical Research Institute, Konyang University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sang-Won Lee
- Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Dokeun Kim
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jung-Sik Yoo
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hye-Sook Jeong
- Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| |
Collapse
|
31
|
An Y, Ni R, Zhuang L, Yang L, Ye Z, Li L, Parkkila S, Aspatwar A, Gong W. Tuberculosis vaccines and therapeutic drug: challenges and future directions. MOLECULAR BIOMEDICINE 2025; 6:4. [PMID: 39841361 PMCID: PMC11754781 DOI: 10.1186/s43556-024-00243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Tuberculosis (TB) remains a prominent global health challenge, with the World Health Organization documenting over 1 million annual fatalities. Despite the deployment of the Bacille Calmette-Guérin (BCG) vaccine and available therapeutic agents, the escalation of drug-resistant Mycobacterium tuberculosis strains underscores the pressing need for more efficacious vaccines and treatments. This review meticulously maps out the contemporary landscape of TB vaccine development, with a focus on antigen identification, clinical trial progress, and the obstacles and future trajectories in vaccine research. We spotlight innovative approaches, such as multi-antigen vaccines and mRNA technology platforms. Furthermore, the review delves into current TB therapeutics, particularly for multidrug-resistant tuberculosis (MDR-TB), exploring promising agents like bedaquiline (BDQ) and delamanid (DLM), as well as the potential of host-directed therapies. The hurdles in TB vaccine and therapeutic development encompass overcoming antigen diversity, enhancing vaccine effectiveness across diverse populations, and advancing novel vaccine platforms. Future initiatives emphasize combinatorial strategies, the development of anti-TB compounds targeting novel pathways, and personalized medicine for TB treatment and prevention. Despite notable advances, persistent challenges such as diagnostic failures and protracted treatment regimens continue to impede progress. This work aims to steer future research endeavors toward groundbreaking TB vaccines and therapeutic agents, providing crucial insights for enhancing TB prevention and treatment strategies.
Collapse
Affiliation(s)
- Yajing An
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ruizi Ni
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Li Zhuang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Ling Yang
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Zhaoyang Ye
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Linsheng Li
- Graduate School, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories PLC, Tampere, Finland
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, 33014, Tampere, Finland.
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, 17#Heishanhu Road, Haidian District, Beijing, 100091, China.
| |
Collapse
|
32
|
Arce-Aceves MF, Espinosa-Neira R, Mata-Espinosa DA, Barrios-Payan JA, Castelán-Sánchez HG, Alcaraz-Estrada SL, Castañón-Arreola M, Hernández-Pando R. Fitness costs of Mycobacterium tuberculosis resistant to rifampicin is compensated by rapid Th2 polarization mediated by early and high IL-4 production during mice infection. Sci Rep 2025; 15:2811. [PMID: 39843896 PMCID: PMC11754857 DOI: 10.1038/s41598-024-81446-3] [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: 07/08/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025] Open
Abstract
It was a general belief that drug resistance in Mycobacterium tuberculosis (Mtb) was associated with lesser virulence, particularly rifampicin resistance, which is usually produced by mutations in the RNA polymerase Beta subunit (RpoB). Interestingly, this kind of bacterial mutations affect gene transcription with significant effects on bacterial physiology and metabolism, affecting also the bacterial antigenic constitution that in consequence can produce diverse immune responses and disease outcome. In the present study, we show the results of the Mtb clinical isolate A96, which is resistant to rifampicin and when used to infect BALB/c mice showed hypervirulence, apparently by rapidly polarization of the Th2 immune response through early and high production of IL-4. The 2D-PAGE analysis of the secretome of Mtb A96 showed 204 spots, and by immunoproteome, seven proteins that were differentially recognized with the sera of infected mice on day 28 were identified by LC-MS/MS. The proteins correspond to surface antigens, virulence factors, and energy metabolism enzymes. Some of them are immunodominant antigens, such as LpqH lipoprotein that induces IL-4 secretion in cell suspensions from the lung and spleen of mice infected with Mtb A96 at 28 days postinfection, suggesting that LpqH could be one of the main antigens involved in the Th2 polarization. The reduction of Mtb A96 hypervirulence in IL-4Rα-/- BALB/c mice highlights the importance of IL-4 induction and Th2 response polarization and the immunopathological response. Thus, high and rapid bias to Th2 response is a mechanism of Mtb virulence, which could be mediated by rifampicin-resistant Mtb isolates, probably by high production and secretion of specific antigens.
Collapse
Affiliation(s)
- Ma Fernanda Arce-Aceves
- Experimental Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Roberto Espinosa-Neira
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290, Colonia Del Valle Sur, Alcaldía Benito Juárez, Ciudad de México, CP. 03100, Mexico
| | - Dulce A Mata-Espinosa
- Experimental Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Jorge A Barrios-Payan
- Experimental Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Hugo G Castelán-Sánchez
- Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 3K7, Canada
| | - Sofía L Alcaraz-Estrada
- Virological Analysis and Reference Unit, Institute for Social Security and Services for State Workers, National Medical Center "20 de Noviembre", Mexico City, Mexico
| | - Mauricio Castañón-Arreola
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290, Colonia Del Valle Sur, Alcaldía Benito Juárez, Ciudad de México, CP. 03100, Mexico.
| | - Rogelio Hernández-Pando
- Experimental Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico.
- Experimental Pathology Department, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Alcaldía Tlalpan, 14080, Ciudad de México, CDMX, Mexico.
| |
Collapse
|
33
|
Faheem I, Nagaraja V. Multifunctional Mycobacterial Topoisomerases with Distinctive Features. ACS Infect Dis 2025. [PMID: 39825760 DOI: 10.1021/acsinfecdis.4c00880] [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: 01/20/2025]
Abstract
Tuberculosis (TB) continues to be a major cause of death worldwide despite having an effective combinatorial therapeutic regimen and vaccine. Being one of the most successful human pathogens, Mycobacterium tuberculosis retains the ability to adapt to diverse intracellular and extracellular environments encountered by it during infection, persistence, and transmission. Designing and developing new therapeutic strategies to counter the emergence of multidrug-resistant and extensively drug-resistant TB remains a major task. DNA topoisomerases make up a unique class of ubiquitous enzymes that ensure steady-state level supercoiling and solve topological problems occurring during DNA transactions in cells. They continue to be attractive targets for the discovery of novel classes of antibacterials and to develop better molecules from existing drugs by virtue of their reaction mechanism. The limited repertoire of topoisomerases in M. tuberculosis, key differences in their properties compared to topoisomerases from other bacteria, their essentiality for the pathogen's survival, and validation as candidates for drug discovery provide an opportunity to exploit them in drug discovery efforts. The present review provides insights into their organization, structure, function, and regulation to further efforts in targeting them for new inhibitor discovery. First, the structure and biochemical properties of DNA gyrase and Topoisomerase I (TopoI) of mycobacteria are described compared to the well-studied counterparts from other bacteria. Next, we provide an overview of known inhibitors of DNA gyrase and emerging novel bacterial topoisomerase inhibitors (NBTIs). We also provide an update on TopoI-specific compounds, highlighting mycobacteria-specific inhibitors.
Collapse
Affiliation(s)
- Iqball Faheem
- Department of Microbiology and Cell Biology, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
| | - Valakunja Nagaraja
- Department of Microbiology and Cell Biology, Indian Institute of Science, C.V. Raman Avenue, Bangalore 560012, India
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
- Indian Institute of Science Education and Research, Bhopal 462066, India
| |
Collapse
|
34
|
Rani N, Rajmani RS, Surolia A. Identification of an Isoxazole Derivative as an Antitubercular Compound for Targeting the FadD Enzymes of Mycobacterium tuberculosis. J Med Chem 2025; 68:270-286. [PMID: 39693602 DOI: 10.1021/acs.jmedchem.4c01844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
FadD32, a fatty acyl-AMP ligase, plays an indispensable role in mycobacterial mycolic acid synthesis and is a validated target for tuberculosis (TB) drug development. The crystal structure of Mycobacterium tuberculosis (Mtb)FadD32 has laid the foundation of structure-based drug discovery against this crucial enzyme. Here, we screened the "isoxazole" scaffold containing molecules against MtbFadD32 and identified a compound 2,4-dibromo-6-[3-(trifluoromethyl)-1,2-oxazol-5-yl]phenol (M1) with specific inhibitory activity against Mtb. Kinetics experiments showed that M1 inhibits MtbFadD32 and MtbFadD28 activity. The transcriptomics response of Mtb disclosed M1-mediated regulation of mycobacterial decisive genes involved in cell wall synthesis, consequently creating unfavorable conditions for Mtb survival. Further, M1 curtails the Mtb survival in infected macrophages and reduces Mtb burden and tubercular granulomas in a chronic infection model of BALB/c mice. Our findings provide an effective chemical scaffold to inhibit MtbFadD32 with the potential to inhibit multiple MtbFadD family of enzymes for further development as a promising candidate for treating TB.
Collapse
Affiliation(s)
- Nidhi Rani
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Raju S Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
- Dr. Reddy's Institute of Life Science, Hyderabad 500046, India
| |
Collapse
|
35
|
Xu D, Gu Y, Li G, Wang R, Xiao S, Duan H, Jiang J, Zhao X, Wan K, He X, Liu H, Lou Y. Evaluation of the cross-immunity between Mycobacterium tuberculosis and Mycobacterium abscessus in vitro. BMC Microbiol 2025; 25:9. [PMID: 39789455 PMCID: PMC11716203 DOI: 10.1186/s12866-024-03724-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Mycobacterium tuberculosis (M. tuberculosis) and Mycobacterium abscessus (M. abscessus) are important pathogens that can cause lung diseases. Given the abundance of shared antigens between these two pathogens, evaluating the cross-immunization between Mycobacterium tuberculosis and Mycobacterium abscessus has implications for the assessment of tuberculosis vaccines based on nontuberculous mycobacteria (NTM). The whole-cell proteins of Mycobacterium abscessus were lysed via ultrasonication and then were subcutaneously injected into BALB/c mice either alone or mixed with adjuvant for three times at a 10-day interval. After the final immunization, cross-immune antigens were analysed via genomic comparison and Mycobacterium tuberculosis proteome microarrays. BALB/c mice splenic lymphocytes were stimulated with TB-PPD to assess the cross-immunity of the cellular immune response. The effect of cross-immunity on the growth of Mycobacterium tuberculosis was evaluated using a Mycobacterium tuberculosis growth inhibition assay. Despite the presence of 1,953 homologous gene clusters between Mycobacterium tuberculosis and Mycobacterium abscessus, only 302 Mycobacterium tuberculosis antigens exhibited cross-immunoreactivity after three immunizations. Compared with the PBS group, TB-PPD stimulation significantly increased the secretion of TNF-α, IL-4, and IL-6 by sensitized mouse splenic lymphocytes, and significantly affected the proliferation of IL-2+CD4 T and TNF-α+CD4 T cells in the immunized group (P < 0.05), but had no impact on IFN-γ and IFN-γ+ CD4 T cells. Furthermore, there was no significant difference in the proliferation of Mycobacterium tuberculosis between the immunized group and the PBS group in spleen cells. These data indicate that proteins from Mycobacterium abscessus are highly immunogenic in mice. However, the cross-immune response between Mycobacterium abscessus and Mycobacterium tuberculosis was inadequate to effectively inhibit the proliferation of Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Da Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yujie Gu
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Guilian Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ruihuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shiqi Xiao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hongyang Duan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jingwei Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiuqin Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kanglin Wan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xinyue He
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Haican Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China.
| | - Yongliang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
36
|
Russell DG, Simwela NV, Mattila JT, Flynn J, Mwandumba HC, Pisu D. How macrophage heterogeneity affects tuberculosis disease and therapy. Nat Rev Immunol 2025:10.1038/s41577-024-01124-3. [PMID: 39774813 DOI: 10.1038/s41577-024-01124-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Macrophages are the primary host cell type for infection by Mycobacterium tuberculosis in vivo. Macrophages are also key immune effector cells that mediate the control of bacterial growth. However, the specific macrophage phenotypes that are required for optimal immune control of M. tuberculosis infection in vivo remain poorly defined. There are two distinct macrophage lineages in the lung, comprising embryonically derived, tissue-resident alveolar macrophages and recruited, blood monocyte-derived interstitial macrophages. Recent studies have shown that these lineages respond divergently to similar immune environments within the tuberculosis granuloma. Here, we discuss how the differing responses of macrophage lineages might affect the control or progression of tuberculosis disease. We suggest that the ability to reprogramme macrophage responses appropriately, through immunological or chemotherapeutic routes, could help to optimize vaccines and drug regimens for tuberculosis.
Collapse
Affiliation(s)
- David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Nelson V Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Henry C Mwandumba
- Malawi Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Davide Pisu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Microbial Pathogenesis and Immunology, Texas A&M School of Medicine, Bryan, TX, USA
| |
Collapse
|
37
|
Li R, He T, Yang M, Xu J, Li Y, Wang X, Guo X, Li M, Xu L. Regulation of Bacillus Calmette-Guérin-induced macrophage autophagy and apoptosis by the AMPK-mTOR-ULK1 pathway. Microbiol Res 2025; 290:127952. [PMID: 39476518 DOI: 10.1016/j.micres.2024.127952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/02/2024] [Accepted: 10/24/2024] [Indexed: 12/12/2024]
Abstract
Tuberculosis (TB) is a chronic wasting infectious disease caused by Mycobacterium tuberculosis (MTB) or Mycobacterium bovis that can be transmitted among people and domestic animals. During the development of TB, macrophages of the innate immune system can act against MTB via autophagy and apoptosis to prevent the spread of the disease. Among the many autophagy regulatory pathways, the adenosine monophosphate (AMP)-activated protein kinase (AMPK)-mammalian rapamycin target protein (mTOR)-Unc-51-like kinase 1 (ULK1) pathway has received considerable attention. This study investigates the regulatory role of the AMPK-mTOR-ULK1 pathway in attenuating M. bovis Bacillus Calmette-Guérin (BCG)-induced autophagy and apoptosis in murine monocyte macrophages (RAW264.7). Changes in macrophage autophagy and apoptosis were analyzed using the AMPK activator AICAR and inhibitor Compound C to interfere with the AMPK-mTOR-ULK1 pathway and siRNA to silence the pathway. Consequently, BCG stimulation of macrophages significantly activated the AMPK-mTOR-ULK1 pathway while BCG-induced macrophage AMPK activation promoted macrophage autophagy and apoptosis. Activation of the AMPK-mTOR-ULK1 pathway by AICAR significantly improved autophagy occurrence in BCG-induced macrophages and increased apoptosis while Compound C with siRNA produced opposing effects by attenuating autophagy and apoptosis in BCG-induced macrophages. Thus, the AMPK-mTOR-ULK1 pathway has a dual regulatory role in BCG-induced macrophage autophagy and apoptosis and may have synergistic effects. This study analyzes the mechanism of resistance of host cells to MTB and provides a theoretical basis for new therapeutic strategies and related drug development.
Collapse
Affiliation(s)
- Ruiqian Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Tianle He
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Min Yang
- Guyuan Vocational and Technical School, Guyuan, Ningxia 756000, China
| | - Jinghua Xu
- COFCO Feed (Yinchuan) Co., Ltd., Lingwu, Ningxia 750499, China
| | - Yongqin Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xueyan Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xuelian Guo
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Mingzhu Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Lihua Xu
- College of Animal Science and Technology, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
38
|
Byrne AS, Bissonnette N, Tahlan K. Mechanisms and implications of phenotypic switching in bacterial pathogens. Can J Microbiol 2025; 71:1-19. [PMID: 39361974 DOI: 10.1139/cjm-2024-0116] [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: 10/05/2024]
Abstract
Bacteria encounter various stressful conditions within a variety of dynamic environments, which they must overcome for survival. One way they achieve this is by developing phenotypic heterogeneity to introduce diversity within their population. Such distinct subpopulations can arise through endogenous fluctuations in regulatory components, wherein bacteria can express diverse phenotypes and switch between them, sometimes in a heritable and reversible manner. This switching may also lead to antigenic variation, enabling pathogenic bacteria to evade the host immune response. Therefore, phenotypic heterogeneity plays a significant role in microbial pathogenesis, immune evasion, antibiotic resistance, host niche tissue establishment, and environmental persistence. This heterogeneity can result from stochastic and responsive switches, as well as various genetic and epigenetic mechanisms. The development of phenotypic heterogeneity may create clonal populations that differ in their level of virulence, contribute to the formation of biofilms, and allow for antibiotic persistence within select morphological variants. This review delves into the current understanding of the molecular switching mechanisms underlying phenotypic heterogeneity, highlighting their roles in establishing infections caused by select bacterial pathogens.
Collapse
Affiliation(s)
| | - Nathalie Bissonnette
- Sherbrooke Research and Development Center, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada
| | - Kapil Tahlan
- Department of Biology, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
39
|
Araújo LE, Petrilli J, Oliveira C, Horta T, Estevão P, Carvalho FR, Cardoso CAA, Cardoso TM, de Ângelis L, Montenegro L, Santos FLN, Arruda S, Queiroz A. Evaluation of nonpolar lipid extract antigen-based enzyme-linked immunosorbent assay for the serodiagnosis of tuberculosis. Diagn Microbiol Infect Dis 2025; 111:116560. [PMID: 39437652 DOI: 10.1016/j.diagmicrobio.2024.116560] [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: 04/04/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
This study assessed the diagnostic potential of nonpolar lipid extracts in enzyme-linked immunosorbent assays (ELISAs) for tuberculosis (TB) serodiagnosis. Nonpolar lipid extracts were harvested from Mycobacterium tuberculosis (Mtb) knockout in mce1 operon (∆mce1) and its parental wild type (WT) strains. IgM and IgG anti-nonpolar lipid serum levels were measured in TB patients (n=45), healthy individuals with positive (n=22) and negative (n=44) interferon-gamma release assay (IGRA) results, and symptomatic respiratory (SR) patients with negative TB tests (n=9). IgG anti-WT lipid distinguished TB patients from IGRA-positive individuals with 60% sensitivity and 77.3% specificity. Conversely, IgG anti-∆mce lipid levels didn't vary significantly across groups. Interestingly, most SR patients exhibited significantly higher IgM and IgG anti-WT lipid titers than the IGRA-positive and -nega groups. While the overall diagnostic potential of Mtb nonpolar lipids was limited, the impaired immunogenecity of Δmce1 lipid extract suggests that some missing lipid classes in this extract can potentially induce antibody production in TB patients.
Collapse
Affiliation(s)
- Luana E Araújo
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Jéssica Petrilli
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Carlos Oliveira
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Thainá Horta
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Paulo Estevão
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Fabiana Rabe Carvalho
- Laboratório Multiusuário de Apoio à Pesquisa em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brasil
| | - Claudete A Araújo Cardoso
- Laboratório Multiusuário de Apoio à Pesquisa em Nefrologia e Ciências Médicas, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brasil; Departamento Materno-Infantil, Faculdade de Medicina, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brasil
| | - Thiago Marconi Cardoso
- Laboratório de Pesquisa Clínica, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Luanna de Ângelis
- Laboratório de Imunoepidemiologia, Instituto Aggeu Magalhães (IAM)/Fiocruz, Recife, Pernambuco, Brasil
| | - Lilian Montenegro
- Laboratório de Imunoepidemiologia, Instituto Aggeu Magalhães (IAM)/Fiocruz, Recife, Pernambuco, Brasil
| | - Fred Luciano Neves Santos
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Sérgio Arruda
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil
| | - Adriano Queiroz
- Laboratório Avançado de Saúde Pública, Instituto Gonçalo Moniz (IGM)/Fiocruz, Salvador, Bahia, Brasil.
| |
Collapse
|
40
|
Hiebert MR, Sharma MK, Go A, Bonner C, Laminman V, Graham M, Soualhine H. RNA extraction and RNA-sequencing method for transcriptomic analysis of Mycobacterium tuberculosis. Biotechniques 2025; 77:23-34. [PMID: 39957238 DOI: 10.1080/07366205.2025.2457887] [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: 08/23/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
RNA-sequencing (RNA-seq) technologies have advanced exponentially in recent years, however, the application of RNA-seq to Mycobacterium tuberculosis remains limited. We present a wet-lab and computational protocol for RNA-seq based transcriptomics that was tested on 12 replicates each of 11 clinical isolates of M. tuberculosis (n = 132) grown in vitro with and without pyrazinamide exposure. This RNA extraction method uses low-volume cultures, mechanical lysis, TRIzol™ phase separation, and column-based purification to produce high yields of pure, intact RNA followed by rRNA depletion and cDNA library preparation. The detection of unique transcripts was optimized at a sequencing depth of 15 million reads. This method detected differential RNA expression in experimental sets with and without pyrazinamide exposure, demonstrating that the method is suitable for RNA-seq applications.
Collapse
Affiliation(s)
- Morgan R Hiebert
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Meenu K Sharma
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Alwyn Go
- Genomics Core Facility, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
| | - Christine Bonner
- Genomics Core Facility, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
| | - Vanessa Laminman
- Genomics Core Facility, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
| | - Morag Graham
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
- Genomics Core Facility, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
| | - Hafid Soualhine
- National Reference Centre for Mycobacteriology, National Microbiology Laboratory Branch, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
41
|
de Souza EV, Dalberto PF, Miranda AC, Saghatelian A, Pinto AM, Basso LA, Machado P, Bizarro CV. Large-scale proteogenomics characterization of microproteins in Mycobacterium tuberculosis. Sci Rep 2024; 14:31186. [PMID: 39732784 DOI: 10.1038/s41598-024-82465-w] [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: 12/07/2023] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Tuberculosis remains a burden to this day, due to the rise of multi and extensively drug-resistant bacterial strains. The genome of Mycobacterium tuberculosis (Mtb) strain H37Rv underwent an annotation process that excluded small Open Reading Frames (smORFs), which encode a class of peptides and small proteins collectively known as microproteins. As a result, there is an overlooked part of its proteome that is a rich source of potentially essential, druggable molecular targets. Here, we employed our recently developed proteogenomics pipeline to identify novel microproteins encoded by non-canonical smORFs in the genome of Mtb using hundreds of mass spectrometry experiments in a large-scale approach. We found protein evidence for hundreds of unannotated microproteins and identified smORFs essential for bacterial survival and involved in bacterial growth and virulence. Moreover, many smORFs are co-expressed and share operons with a myriad of biologically relevant genes and play a role in antibiotic response. Together, our data presents a resource of unknown genes that play a role in the success of Mtb as a widespread pathogen.
Collapse
Affiliation(s)
- Eduardo V de Souza
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Pedro F Dalberto
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
| | - Adriana C Miranda
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Antonio M Pinto
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Luiz A Basso
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
| | - Pablo Machado
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil
| | - Cristiano V Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional (CPBMF) and Instituto Nacional de Ciência e Tecnologia em Tuberculose (INCT-TB), Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil.
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, 90619-900, Brazil.
| |
Collapse
|
42
|
Kalinich CC, Gonzalez FL, Osmaston A, Breban MI, Distefano I, Leon C, Sheen P, Zimic M, Coronel J, Tan G, Crudu V, Ciobanu N, Codreanu A, Solano W, Ráez J, Allicock OM, Chaguza C, Wyllie AL, Brandt M, Weinberger DM, Sobkowiak B, Cohen T, Grandjean L, Grubaugh ND, Redmond SN. Tiled Amplicon Sequencing Enables Culture-free Whole-Genome Sequencing of Pathogenic Bacteria From Clinical Specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629550. [PMID: 39763738 PMCID: PMC11702625 DOI: 10.1101/2024.12.19.629550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Pathogen sequencing is an important tool for disease surveillance and demonstrated its high value during the COVID-19 pandemic. Viral sequencing during the pandemic allowed us to track disease spread, quickly identify new variants, and guide the development of vaccines. Tiled amplicon sequencing, in which a panel of primers is used for multiplex amplification of fragments across an entire genome, was the cornerstone of SARS-CoV-2 sequencing. The speed, reliability, and cost-effectiveness of this method led to its implementation in academic and public health laboratories across the world and adaptation to a broad range of viral pathogens. However, similar methods are not available for larger bacterial genomes, for which whole-genome sequencing typically requires in vitro culture. This increases costs, error rates and turnaround times. The need to culture poses particular problems for medically important bacteria such as Mycobacterium tuberculosis, which are slow to grow and challenging to culture. As a proof of concept, we developed two novel whole-genome amplicon panels for M. tuberculosis and Streptococcus pneumoniae. Applying our amplicon panels to clinical samples, we show the ability to classify pathogen subgroups and to reliably identify markers of drug resistance without culturing. Development of this work in clinical settings has the potential to dramatically reduce the time of diagnosis of drug resistance for multiple drugs in parallel, enabling earlier intervention for high priority pathogens.
Collapse
Affiliation(s)
- Chaney C Kalinich
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Freddy L Gonzalez
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
| | - Alice Osmaston
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mallery I Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Isabel Distefano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Candy Leon
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Mirko Zimic
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Grace Tan
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
| | | | | | | | | | - Jimena Ráez
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Orchid M Allicock
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
| | - Chrispin Chaguza
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anne L Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Matthew Brandt
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Benjamin Sobkowiak
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ted Cohen
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Louis Grandjean
- Department of Infection, Immunity, and Inflammation, Institute of Child Health, University College Longon, London, England
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nathan D Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
- Public Health Modeling Unit, Yale School of Public Health, New Haven, Connecticut, USA
| | - Seth N Redmond
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
43
|
Dahiya P, Banerjee A, Saha A, Nandicoori VK, Ghosh S, Mukhopadhyay S. Structure-function relationship of PE11 esterase of Mycobacterium tuberculosis with respect to its role in virulence. Biochem Biophys Res Commun 2024; 739:150927. [PMID: 39541926 DOI: 10.1016/j.bbrc.2024.150927] [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: 07/12/2024] [Revised: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
The lipolytic enzymes of Mycobacterium tuberculosis play a critical role in immunomodulation and virulence. Among these proteins, PE11 which also belongs to the PE/PPE family, is the smallest (∼10.8 kDa) and play a significant role in cell wall remodelling and virulence. PE11 is established to be an esterase, but its enzymatic and structural properties are not yet characterized. In this study, using homology modelling we deduced the putative structure which shows the presence of both α-helix and β-sheet structures which is in close agreement with that observed by CD spectra of the purified protein. PE11 was found to contain a Gx3Sx4G motif homologous to canonical 'GxSxG' motif present in many serin hydrolases. The catalytic triad appears to be located within this motif as substitution of Serine26 and Glycine31 residues abrogated its enzymatic activity. Gel-filtration chromatography data indicate that PE11 possibly exists as dimer and tetramer showing positive cooperativity for binding its substrates. In addition, PE11 esterase activity was found to be critical for cell wall remodelling, antibiotic resistance and conferring survival advantages to M. tuberculosis. Our data suggest that PE11 can be targeted for designing potential therapeutic strategies.
Collapse
Affiliation(s)
- Priyanka Dahiya
- Laboratory of Molecular Cell Biology, BRIC-Center for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, 500039, Telangana, India; Graduate Studies, Regional Center for Biotechnology, Haryana, India
| | - Amit Banerjee
- ICMR-National Institute of Nutrition, Hyderabad, 500007, Telangana, India
| | - Abhishek Saha
- Centre for Cellular and Molecular Biology, Hyderabad, 500007, Telangana, India
| | | | - Sudip Ghosh
- ICMR-National Institute of Nutrition, Hyderabad, 500007, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, BRIC-Center for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, 500039, Telangana, India.
| |
Collapse
|
44
|
Lin S, Xu F, Huang B, Zhao LL, Pan D, Lin S. Visual codon: a user-friendly Python program for viewing and optimizing gene GC content. PeerJ 2024; 12:e18755. [PMID: 39717051 PMCID: PMC11665431 DOI: 10.7717/peerj.18755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024] Open
Abstract
Due to the codon bias of different species, codon optimization is usually carried out in the process of heterologous protein expression. At present, there are a variety of codon optimization tools. However, the optimized sequences may still have high or low points of local guanine and cytosine (GC) content, which is not conducive to the primer design of gene subcloning, and also makes it difficult to perform the experiment of synthesizing the whole gene with DNA fragments by polymerase chain reaction (PCR) reaction. In this study, we present a stand-alone software written in Python, with which users can manually check and adjust the GC content of sequence-optimized genes. The software takes the codon frequency of Escherichia coli as default and can work with other species as well. It provides a Graphical User Interface (GUI) interface, which allows users to change codons and intuitively see the effect of codon changes on local GC content. Our program brings convenience for the optimization of gene GC content and the subsequent gene cloning experiments.
Collapse
Affiliation(s)
- Shiming Lin
- School of Computing and Information Science, Fuzhou Institute of Technology, Fuzhou, Fujian, China
| | - Fei Xu
- College of Agronomy, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Bifang Huang
- Life Science College, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Li-li Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Danni Pan
- College of Agronomy, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Shiqiang Lin
- Life Science College, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| |
Collapse
|
45
|
Kokot M, Hrast Rambaher M, Feng L, Mitchenall LA, Lawson DM, Maxwell A, Parish T, Minovski N, Anderluh M. Structural Aspects of Mycobacterium tuberculosis DNA Gyrase Targeted by Novel Bacterial Topoisomerase Inhibitors. ACS Med Chem Lett 2024; 15:2164-2170. [PMID: 39691510 PMCID: PMC11647683 DOI: 10.1021/acsmedchemlett.4c00447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
In this Letter, we present a small series of novel bacterial topoisomerase inhibitors (NTBIs) that exhibit both potent inhibition of Mycobacterium tuberculosis DNA gyrase and potent antimycobacterial activity. The disclosed crystal structure of M. tuberculosis DNA gyrase in complex with DNA and compound 5 from this NBTI series reveals the binding mode of an NBTI in the GyrA binding pocket and confirms the presence and importance of halogen bonding for the excellent on-target potency. In addition, we have shown that compound 5 is a promising M. tuberculosis DNA gyrase inhibitor, with an IC50 for M. tuberculosis gyrase of 0.096 μM, and it has potent activity against M. tuberculosis, with an IC50 of 0.165 μM.
Collapse
Affiliation(s)
- Maja Kokot
- Theory
Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Martina Hrast Rambaher
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Lipeng Feng
- Department
of Biological Chemistry, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
- Department
of Molecular Microbiology, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Lesley A Mitchenall
- Department
of Biological Chemistry, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
- Department
of Molecular Microbiology, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
| | - David M Lawson
- Department
of Biological Chemistry, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
- Department
of Biochemistry & Metabolism, John Innes
Centre, Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Anthony Maxwell
- Department
of Biological Chemistry, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
- Department
of Molecular Microbiology, John Innes Centre,
Norwich Research Park, Norwich NR4 7UH, U.K.
| | - Tanya Parish
- School of
Medicine, University of Washington, Seattle, Washington 98195, United States
- Center
for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington 98105, United States
| | - Nikola Minovski
- Theory
Department, Laboratory for Cheminformatics, National Institute of Chemistry, Hajdrihova 19, 1001 Ljubljana, Slovenia
| | - Marko Anderluh
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
46
|
Maharjan R, Zhang Z, Klenotic PA, Gregor WD, Purdy GE, Yu EW. Cryo-EM structure of the Mycobacterium smegmatis MmpL5-AcpM complex. mBio 2024; 15:e0303524. [PMID: 39480109 PMCID: PMC11633376 DOI: 10.1128/mbio.03035-24] [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: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Mycobacterium tuberculosis, the causative agent of the airborne infection tuberculosis (TB), contains 13 mycobacterial membrane protein large (MmpL) transporters that can be divided into two distinct subclasses. These MmpL proteins play important functional roles within the mycobacterium and subsequently are considered attractive drug targets to combat TB infection. Previously, we reported both X-ray and cryo-electron microscopy (cryo-EM) structures of the MmpL3 transporter, providing high-resolution structural information for this subclass of the MmpL proteins. Thus far, there is no structural information available for the other subclass, which includes MmpL5, an inner membrane transporter that plays a critical role in iron hemostasis. Here, we report the first cryo-EM structure of the Mycobacterium smegmatis MmpL5 transporter bound with the meromycolate extension acyl carrier protein M (AcpM) to a resolution of 2.81 Å. Our structural data reveals that MmpL5 and AcpM interact in the cytoplasm to form a complex, and this allows us to propose that MmpL5 may also associate with the mycobactin L (MbtL) protein in a similar fashion to form a heterocomplex important for iron acquisition, which enables the survival and replication of the mycobacterium. IMPORTANCE The emergence and spread of multidrug-resistant tuberculosis (TB) present enormous challenges to the global public health. The causative agent, Mycobacterium tuberculosis, has now infected more than one-third of the world's population. Here, we report the first structure of the mycobacterial membrane protein large 5 (MmpL5), an essential transporter for iron acquisition, bound with the meromycolate extension acyl carrier protein M (AcpM), indicating a plausible pathway for mycobactin translocation. Our studies will ultimately inform an era in structure-guided drug design to combat TB infection.
Collapse
Affiliation(s)
- Rakesh Maharjan
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Zhemin Zhang
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Philip A. Klenotic
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - William D. Gregor
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Georgiana E. Purdy
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Edward W. Yu
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Chatterjee D, Daya Manasi AR, Rastogi SK, Panda AP, Biju B, Bhattacharyya D, Ghosh AS. Involvement of CorA of Mycobacterium smegmatis in exerting intrinsic resistance towards structurally unrelated antibiotics. J Appl Microbiol 2024; 135:lxae298. [PMID: 39657998 DOI: 10.1093/jambio/lxae298] [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: 08/14/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
AIM Ubiquitous magnesium transporter, CorA of Mycobacterium smegmatis is well known for its role in maintaining magnesium homeostasis. However, little is known about its involvement in exerting antimicrobial resistance. Here, by using molecular genetics, in vivo and in silico studies, we tried to envisage the role of CorA of M. smegmatis in antimicrobial resistance of M. smegmatis and Escherichia coli. METHODS AND RESULTS Expression of corA in M. smegmatis and E. coli decreased the susceptibility of the host cells towards various antibiotics and anti-tubercular drugs, which was elucidated by determining minimum inhibitory concentrations using the micro-broth dilution method. The intracellular antibiotic accumulation assay indicated that the host cells expressing corA accumulated less EtBr, norfloxacin, and ofloxacin than the control cells. Moreover, the presence of a sub-inhibitory concentration of Mg2+ further decreased the susceptibility towards the drugs tested. Furthermore, CorA enhanced the biofilm-forming ability of cells expressing it. CONCLUSION CorA (MSMEG_5056), a magnesium transporter of M. smegmatis influences the extrusion of multiple structurally unrelated classes of drugs and enhances the biofilm formation of E. coli and M. smegmatis.
Collapse
Affiliation(s)
- Debasmita Chatterjee
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - A R Daya Manasi
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Sumit Kumar Rastogi
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Aditya Prasad Panda
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Bayomi Biju
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Debleena Bhattacharyya
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| | - Anindya Sundar Ghosh
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal 721302, India
| |
Collapse
|
48
|
Tang W, Gui C, Zhang T. Expression, Purification, and Bioinformatic Prediction of Mycobacterium tuberculosis Rv0439c as a Potential NADP +-Retinol Dehydrogenase. Mol Biotechnol 2024; 66:3559-3572. [PMID: 37989944 DOI: 10.1007/s12033-023-00956-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023]
Abstract
Although the genome of Mycobacterium tuberculosis (Mtb) H37Rv, the causative agent of tuberculosis, has been repeatedly annotated and updated, a range of proteins from this human pathogen have unknown functions. Mtb Rv0439c, a member of the short-chain dehydrogenase/reductases superfamily, has yet to be cloned and characterized, and its function remains unclear. In this work, we present for the first time the optimized expression and purification of this enzyme, as well as bioinformatic analysis to unveil its potential coenzyme and substrate. Optimized expression in Escherichia coli yielded soluble Rv0439c, while certain tag fusions resulted in insolubility. Sequence and docking analyses strongly suggested that Rv0439c has a clear preference for NADP+, with Arg53 being a key residue that confers coenzyme specificity. Furthermore, functional prediction using CLEAN and DEEPre servers suggested that this protein is a potential NADP+-retinol dehydrogenase (EC No. 1.1.1.300) in retinol metabolism, and this was supported by a BLASTp search and docking studies. Collectively, our findings provide a solid basis for future functional characterization and structural studies of Rv0439c, which will contribute to enhanced understanding of Mtb biology.
Collapse
Affiliation(s)
- Wanggang Tang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui, China.
| | - Chuanyue Gui
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China
- School of Public Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Tingting Zhang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China
- School of Public Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| |
Collapse
|
49
|
Megalizzi V, Tanina A, Grosse C, Mirgaux M, Legrand P, Dias Mirandela G, Wohlkönig A, Bifani P, Wintjens R. Domain architecture of the Mycobacterium tuberculosis MabR ( Rv2242), a member of the PucR transcription factor family. Heliyon 2024; 10:e40494. [PMID: 39641026 PMCID: PMC11617747 DOI: 10.1016/j.heliyon.2024.e40494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/28/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
MabR (Rv2242), a PucR-type transcription factor, plays a crucial role in regulating mycolic acid biosynthesis in Mycobacterium tuberculosis. To understand its regulatory mechanisms, we determined the crystal structures of its N-terminal and C-terminal domains. The N-terminal domain adopts a globin-like fold, while the C-terminal domain comprises an α/β GGDEF domain and an all-α effector domain with a helix-turn-helix DNA-binding motif. This unique domain combination is specific to Actinomycetes. Biochemical and computational studies suggest that full-length MabR forms both dimeric and tetrameric assemblies in solution. Structural analysis revealed two distinct dimerization interfaces within the N- and C-terminal domains, further supporting a tetrameric organization. These findings provide valuable insights into the domain architecture, oligomeric state, and potential regulatory mechanisms of MabR.
Collapse
Affiliation(s)
- Véronique Megalizzi
- Unit of Microbiology, Bioorganic and Macromolecular Chemistry, Department of Research in Drug Development, Faculty of Pharmacy, Université Libre de Bruxelles, Belgium
| | - Abdalkarim Tanina
- Unit of Microbiology, Bioorganic and Macromolecular Chemistry, Department of Research in Drug Development, Faculty of Pharmacy, Université Libre de Bruxelles, Belgium
| | - Camille Grosse
- Unit of Microbiology, Bioorganic and Macromolecular Chemistry, Department of Research in Drug Development, Faculty of Pharmacy, Université Libre de Bruxelles, Belgium
| | - Manon Mirgaux
- Unit of Microbiology, Bioorganic and Macromolecular Chemistry, Department of Research in Drug Development, Faculty of Pharmacy, Université Libre de Bruxelles, Belgium
- Laboratoire de Chimie Biologique Structurale (CBS), Unité de Chimie Physique Théorique et Structurale (UCPTS), Department of Chemistry, Faculty of Sciences, University of Namur, Belgium
- Center of Microscopy and Molecular Imaging (CMMI), Biopark Charleroi, Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Gaëtan Dias Mirandela
- Biology of Membrane Transport Laboratory, Molecular Biology Department, Faculty of Sciences, Université Libre de Bruxelles, Belgium
| | - Alexandre Wohlkönig
- Center for Structural Biology, Vlaams Institute voor Biotechnology (VIB), Brussels, Belgium
| | - Pablo Bifani
- A∗STAR Infectious Diseases Laboratory, Agency for Science, Technology and Research (A∗STAR), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - René Wintjens
- Unit of Microbiology, Bioorganic and Macromolecular Chemistry, Department of Research in Drug Development, Faculty of Pharmacy, Université Libre de Bruxelles, Belgium
| |
Collapse
|
50
|
Grijsen ML, Nguyen TH, Pinheiro RO, Singh P, Lambert SM, Walker SL, Geluk A. Leprosy. Nat Rev Dis Primers 2024; 10:90. [PMID: 39609422 DOI: 10.1038/s41572-024-00575-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 11/30/2024]
Abstract
Leprosy, a neglected tropical disease, causes significant morbidity in marginalized communities. Before the COVID-19 pandemic, annual new case detection plateaued for over a decade at ~200,000 new cases. The clinical phenotypes of leprosy strongly parallel host immunity to its causative agents Mycobacterium leprae and Mycobacterium lepromatosis. The resulting spectrum spans from paucibacillary leprosy, characterized by vigorous pro-inflammatory immunity with few bacteria, to multibacillary leprosy, harbouring large numbers of bacteria with high levels of seemingly non-protective, anti-M. leprae antibodies. Leprosy diagnosis remains clinical, leaving asymptomatic individuals with infection undetected. Antimicrobial treatment is effective with recommended multidrug therapy for 6 months for paucibacillary leprosy and 12 months for multibacillary leprosy. The incubation period ranges from 2 to 6 years, although longer periods have been described. Given this lengthy incubation period and dwindling clinical expertise, there is an urgent need to create innovative, low-complexity diagnostic tools for detection of M. leprae infection. Such advancements are vital for enabling swift therapeutic and preventive interventions, ultimately transforming patient outcomes. National health-care programmes should prioritize early case detection and consider post-exposure prophylaxis for individuals in close contact with affected persons. These measures will help interrupt transmission, prevent disease progression, and mitigate the risk of nerve damage and disabilities to achieve the WHO goal 'Towards Zero Leprosy' and reduce the burden of leprosy.
Collapse
Affiliation(s)
- Marlous L Grijsen
- Oxford University Clinical Research Unit Indonesia, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Thuan H Nguyen
- University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Pushpendra Singh
- Microbial Pathogenesis & Genomics Laboratory, ICMR-National Institute of Research in Tribal Health, Jabalpur, Madhya Pradesh, India
| | - Saba M Lambert
- London School of Hygiene & Tropical Medicine, Faculty of Infectious Diseases, London, UK
- Africa Leprosy, Tuberculosis, Rehabilitation and Training (ALERT) Hospital, Addis Ababa, Ethiopia
| | - Stephen L Walker
- London School of Hygiene & Tropical Medicine, Faculty of Infectious Diseases, London, UK
| | - Annemieke Geluk
- Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|