1
|
Tanriver G, Ali Khan S, Góra A, Chegou NN, Mahmoudi S. Exploring the multifaceted roles of resuscitation-promoting factors in tuberculosis: Implications for diagnosis, vaccine development, and drug targeting. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 46:e00886. [PMID: 40201451 PMCID: PMC11978375 DOI: 10.1016/j.btre.2025.e00886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Tuberculosis (TB) remains a significant global health challenge, necessitating continued research efforts to unravel its complex pathogenesis and advance diagnostic, therapeutic, and preventive strategies. Resuscitation-promoting factors (Rpfs) are peptidoglycan-hydrolyzing enzymes that have gained attention due to their key roles in TB infection dynamics. This review aims to provide a comprehensive overview of Rpfs in TB, highlighting their immunological roles, diagnostic potential, and implications for vaccine development and drug targeting through both in silico and experimental approaches. Rpfs exhibit diverse roles in TB, influencing bacterial resuscitation from dormancy, and immune modulation through interactions with host immune cells, such as dendritic cells, and they are potential targets for novel diagnostic and therapeutic interventions. Their ability to stimulate immune responses, particularly interferon-γ production by T cells, underscores their potential as vaccine candidates against TB. Moreover, Rpfs represent promising targets for drug discovery, with inhibitors potentially disrupting bacterial resuscitation and growth.
Collapse
Affiliation(s)
- Gamze Tanriver
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, Gliwice 44-100, Poland
| | - Salman Ali Khan
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, Gliwice 44-100, Poland
| | - Artur Góra
- Tunneling Group, Biotechnology Centre, Silesian University of Technology, Gliwice 44-100, Poland
| | - Novel N Chegou
- South African Medical Research Council Centre for Tuberculosis Research, Division of Immunology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town 8000, South Africa
| | - Shima Mahmoudi
- Biotechnology Centre, Silesian University of Technology, Krzywoustego 8, Gliwice 44-100, Poland
| |
Collapse
|
2
|
Powell SM, Dessi N, Bennett M, Wang B, Robertson A, Waller E, Smith GC, Delahay RJ. Tuberculosis in found dead badgers at the edge of the expanding bovine tuberculosis epidemic. Sci Rep 2025; 15:10547. [PMID: 40148383 PMCID: PMC11950293 DOI: 10.1038/s41598-025-86930-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/15/2025] [Indexed: 03/29/2025] Open
Abstract
Bovine tuberculosis (bTB) is a major disease of cattle in the UK, placing a significant economic burden on the taxpayer. The causative agent, Mycobacterium bovis, has a wide host range, including the European badger (Meles meles). While badgers have been implicated in the transmission and maintenance of infection in cattle in areas of endemic disease, their role at the edge of the endemic area is poorly understood. Here we present data on the prevalence of infection in badgers collected along the southern edge of England's bTB epidemic. Stakeholders across five counties (Oxfordshire, Berkshire, Buckinghamshire, Hampshire, and East Sussex) submitted found-dead badgers for post-mortem examination and testing by bacterial culture. The overall prevalence, as confirmed by whole genome sequencing, was 6.5% (28/428), ranging between 1.1% (1/88) in Hampshire and 13.0% (14/108) in Oxfordshire. The commonest M. bovis clade in badgers was B6-62, which was predominant in 4/5 counties. B6-62 was also the commonest clade found in cattle and was detected in all counties except East Sussex where, although absent from the cattle population, it was detected in local badgers. This study highlights the co-incidence of infection in badgers and cattle in parts of the southern edge area consistent with localised clustering of infection in both species.
Collapse
Affiliation(s)
- Siân M Powell
- National Wildlife Management Centre, Animal and Plant Health Agency, Stonehouse, GL10 3UJ, UK.
| | - Nicola Dessi
- National Wildlife Management Centre, Animal and Plant Health Agency, Stonehouse, GL10 3UJ, UK
| | - Malcolm Bennett
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, LE12 5RD, UK
| | - Belinda Wang
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, LE12 5RD, UK
| | - Andrew Robertson
- Department for Environment, Food and Rural Affairs, 1a Page Street, London, SW1P 4PQ, UK
| | - Elisabeth Waller
- Department of Epidemiological Sciences, Animal and Plant Health Agency, Weybridge, Addlestone, KT15 3NB, UK
| | - Graham C Smith
- National Wildlife Management Centre, Animal and Plant Health Agency, Stonehouse, GL10 3UJ, UK
| | - Richard J Delahay
- National Wildlife Management Centre, Animal and Plant Health Agency, Stonehouse, GL10 3UJ, UK
| |
Collapse
|
3
|
Rabodoarivelo MS, Hoffmann E, Gaudin C, Aguilar-Ayala DA, Galizia J, Sonnenkalb L, Dal Molin M, Cioetto-Mazzabò L, Degiacomi G, Recchia D, Rybniker J, Manganelli R, Pasca MR, Ramón-García S, Lucía A. Protocol to quantify bacterial burden in time-kill assays using colony-forming units and most probable number readouts for Mycobacterium tuberculosis. STAR Protoc 2025; 6:103643. [PMID: 40067825 PMCID: PMC11931380 DOI: 10.1016/j.xpro.2025.103643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/26/2024] [Accepted: 01/31/2025] [Indexed: 03/26/2025] Open
Abstract
Here, we present a protocol to perform a time-kill assay (TKA) to quantify bacterial burden at multiple time points using colony-forming units and most probable number readouts simultaneously. We describe steps for preparing inoculum, experimental conditions, and sampling bacterial counts. We then detail procedures for quantification and analysis. TKAs provide longitudinal data reflecting the dynamics of the antibiotic effect over time against a planktonic culture and quantify the concentration-effect relationship. For complete details on the use and execution of this protocol, please refer to Van Wijk et al.1.
Collapse
Affiliation(s)
| | - Eik Hoffmann
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| | - Cyril Gaudin
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| | | | - Jordana Galizia
- Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Lindsay Sonnenkalb
- Molecular and Experimental Mycobacteriology, Research Center Borstel Leibniz Lung Center, Borstel, Germany
| | - Michael Dal Molin
- Department I of Internal Medicine, Division of Infectious Diseases, University of Cologne, Cologne, Germany; Faculty of Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | | | - Giulia Degiacomi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Deborah Recchia
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Jan Rybniker
- Department I of Internal Medicine, Division of Infectious Diseases, University of Cologne, Cologne, Germany; Faculty of Medicine, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | | | - Maria Rosalia Pasca
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Santiago Ramón-García
- Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, Zaragoza, Spain; CIBER Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Research & Development Agency of Aragon Foundation (Fundación ARAID), Zaragoza, Spain
| | - Ainhoa Lucía
- Department of Microbiology, Pediatrics, Radiology and Public Health, University of Zaragoza, Zaragoza, Spain; CIBER Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Reyes-González LV, Hernández de la Cruz ON, Castañón-Arreola M. Expression of Resuscitation-Promoting Factor C Stimulates the Growth of Mycobacterium bovis BCG and Delays DevR Regulon Activation in Hypoxia. Int J Microbiol 2025; 2025:2139933. [PMID: 39996126 PMCID: PMC11850072 DOI: 10.1155/ijm/2139933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Latent tuberculosis is characterized by the presence of dormant, nonreplicating (DNR) bacilli for years without causing clinical signs and symptoms, remaining as a major reservoir for active tuberculosis. The mechanism through which M. tuberculosis transits from DNR to active bacilli remains unclear. However, resuscitation-promoting factors (Rpfs) could participate in the reactivation. Using recombinant M. bovis BCG that expresses rpfC (M. bovis BCG-pMV261::rpfC), we evaluated the role of RpfC in the growth of bacilli and the expression of 11 hypoxia-regulated genes in comparison with M. bovis BCG-pMV261. The strains were grown in normoxic (21% O2), hypoxic (5% O2), and anoxic (< 0.1% O2) conditions. In normoxic culture, M. bovis BCG-pMV261::rpfC displays a lower expression of sigB and fdxA. In anoxic culture, we did not observe drastic changes in the gene expression, except for those involved in electron transport during anaerobic respiration (pdxA, pfkB, and nark2), whose expression was significantly lower in M. bovis BCG-pMV261. When the strains were cultured in hypoxia, significantly higher culturability was observed in M. bovis BCG-pMV261::rpfC compared to M. bovis BCG-pMV261. This response was accompanied by a higher sigB and sigE expression. In both strains, we observed a higher dosT, devR, fdxA, and fpkB expression in response to hypoxia. Interestingly, except for fdxA, the expression of these genes was lower in M. bovis BCG-pMV261::rpfC. The protein profiles of M. bovis BCG-pMV261::rpfC reflected the maintenance of an active replicative state (similar to that of the strain grown in normoxic conditions). In anoxic cultures, no significant changes were observed in the expression of hypoxia-response genes. These findings suggest that rpfC may have a significant physiological role in inducing the growth of M. bovis BCG-pMV261::rpfC, which results in the delayed activation of genes related to the transition to anaerobic metabolism.
Collapse
|
5
|
Neumann M, Reimann M, Chesov D, Popa C, Dragomir A, Popescu O, Munteanu R, Hölscher A, Honeyborne I, Heyckendorf J, Lange C, Hölscher C, Kalsdorf B. The molecular bacterial load assay predicts treatment responses in patients with pre-XDR/XDR-tuberculosis more accurately than GeneXpert Ultra MTB/Rif. J Infect 2025; 90:106399. [PMID: 39733827 DOI: 10.1016/j.jinf.2024.106399] [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: 07/05/2024] [Revised: 10/13/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
OBJECTIVES Early detection of treatment failure is essential to improve the management of drug-resistant tuberculosis (DR-TB). We evaluated the molecular bacterial load assay (MBLA) in comparison to standard diagnostic tests for monitoring therapy of patients affected by drug-resistant TB. METHODS The performance of MBLA in tracking treatment response in a prospective cohort of patients with pulmonary MDR/RR- and pre-XDR/XDR-TB was compared with mycobacterial culture, mycobacterial DNA detection using GeneXpert (Xpert) and microscopy detection of sputum acid-fast-bacilli. RESULTS Mycobacterium tuberculosis culture conversion was used as the read-out for treatment responses. The MBLA was most concordant during the early phase of treatment, detecting changes in bacterial load with similar accuracy to microscopy and outperforming Xpert. When considering all timepoints, concordance with MGIT results was 72.1% for MBLA, 57.4% for Xpert and 76.7% for microscopy. The AUC for culture conversion was higher for MBLA (0.88, CI 0.84-0.95) than for Xpert (0.78, CI 0.72-0.85) and microscopy (0.77, CI 0.71-0.83). CONCLUSIONS MBLA was superior in the early identification of successful culture conversion compared to microscopy and Xpert and could be a useful biomarker to evaluate novel entities in Phase IIA early-bactericidal-activity drug trials regardless of the degree of M. tuberculosis drug resistance.
Collapse
Affiliation(s)
- Marit Neumann
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| | - Maja Reimann
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany
| | - Dumitru Chesov
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Nicolae Testemitanu State University of Medicine and Pharmacy, Stefan cel Mare si Sfant Boulevard 165, MD-2004 Chisinau, Republic of Moldova
| | - Cristina Popa
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Antonela Dragomir
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Oana Popescu
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Roxana Munteanu
- National Institute of Pneumology Marius Nasta, Șoseaua Viilor 90, 050159 Bucharest, Romania
| | - Alexandra Hölscher
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany
| | - Isobella Honeyborne
- Centre for Clinical Microbiology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Jan Heyckendorf
- Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Internal Medicine I, University Hospitals Schleswig-Holstein, Kiel, Germany; Pulmonology and Inflammatory Medicine, Christian Albrechts-University, Kiel, Germany
| | - Christoph Lange
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany; Baylor College of Medicine and Texas Children´s Hospital, Houston, TX, USA
| | - Christoph Hölscher
- Division of Infection Immunology, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany; German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany.
| | - Barbara Kalsdorf
- German Center for Infection Research (DZIF), Partner site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Division of Clinical Infectious Diseases, Research Center Borstel, Parkallee 1-40, 23845 Borstel, Germany
| |
Collapse
|
6
|
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
|
7
|
Shleeva MO, Demina GR, Kaprelyants AS. Biochemistry of Reactivation of Dormant Mycobacteria. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S193-S213. [PMID: 40164159 DOI: 10.1134/s0006297924603757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/15/2024] [Accepted: 08/23/2024] [Indexed: 04/02/2025]
Abstract
An important aspect of medical microbiology is identification of the causes and mechanisms of reactivation (resuscitation) of dormant non-sporulating bacteria. In particular, dormant Mycobacterium tuberculosis (Mtb) can cause latent tuberculosis (TB), which could be reactivated in the human organism to the active form of the disease. Analysis of experimental data suggested that reactivation of mycobacteria and reversion of the growth processes include several stages. The initial stage is associated with breakdown of the storage substances like trehalose upon the action of trehalase and with peptidoglycan hydrolysis. Demethylation of tetramethyl porphyrins accumulated in hydrophobic sites (membranes) of the dormant cell also occur in this stage. Metabolic reactivation, starting with cAMP synthesis and subsequent activation of metabolic reactions and biosynthetic processes take place at the stage two as has been shown in the omics studies. Mechanisms of cell reactivation by exogenous free fatty acids via activation of adenylate cyclase and cAMP production have been also suggested. Onset of the cell division is a key benchmark of the third and final stage. Hydrolysis of peptidoglycan as a result of enzymatic action of peptidoglycan hydrolases of the Rpf family is an important process in reactivation of the dormant mycobacteria. Two possible mechanisms for participation of Rpf proteins in reactivation of the dormant bacteria are discussed. On the one hand, muropeptides could be formed as products of peptidoglycan hydrolysis, which could interact with appropriate receptors in bacterial cells transducing activating signal via the PknB phosphotransferase. On the other hand, Rpf protein could presumably change structure of the cell wall, making it more permeable to nutrients and substrates. Both hypotheses were examined in this review. Upon reactivation, independent enzymatic reactions resume their functioning from the beginning of reactivation. Such activation of the entire metabolism occurs rather stochastically, which concludes in combining all biochemical processes in one. This review presents current knowledge regarding biochemical mechanisms of the dormant mycobacteria reactivation, which is important for both fundamental and medical microbiology.
Collapse
Affiliation(s)
- Margarita O Shleeva
- Federal Research Centre "Fundamentals of Biotechnology", A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow, 119071, Russia.
| | - Galina R Demina
- Federal Research Centre "Fundamentals of Biotechnology", A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Arseny S Kaprelyants
- Federal Research Centre "Fundamentals of Biotechnology", A. N. Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow, 119071, Russia
| |
Collapse
|
8
|
Maitra A, Wijk M, Margaryan H, Denti P, McHugh TD, Kloprogge F. The impact of physiological state and environmental stress on bacterial load estimation methodologies for Mycobacterium tuberculosis. Sci Rep 2024; 14:26108. [PMID: 39477982 PMCID: PMC11525806 DOI: 10.1038/s41598-024-74318-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
When processed in solid or liquid medium, tuberculosis patient samples yield different proportions of a heterogenous bacterial community over the duration of treatment. We aimed to derive a relationship between methodologies for bacterial load determination and assess the effect of the growth phase of the parent culture and its exposure to stress on the results. Mycobacterium tuberculosis H37Rv was grown with and without antibiotic (isoniazid or rifampicin) and sampled on day 0, 3, 11 and 21 of growth in broth culture. The bacterial load was estimated by colony counts and the BD BACTEC MGIT system. Linear and nonlinear mixed-effects models were used to describe the relationship between time-to-positivity (TTP) and time-to-growth (TTG) versus colony forming units (CFU), and growth units (GU) versus incubation time in MGIT. For samples with the same CFU, antibiotic-treated and stationary phase cells had a shorter TTP than antibiotic-free controls and early-logarithmic phase cells, respectively. Similarly, stationary phase samples reached higher GUs and had shorter TTG than early-log phase ones. This suggests that there is a population of bacterial cells that can be differentially recovered in liquid medium, giving us insight into the physiological states of the original culture, aiding the interpretation of clinical trial outputs.
Collapse
Affiliation(s)
- Arundhati Maitra
- Institute for Global Health, University College London, London, UK
- Centre for Clinical Microbiology, University College London, London, UK
| | - Marie Wijk
- Institute for Global Health, University College London, London, UK.
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa.
| | - Hasmik Margaryan
- Centre for Clinical Microbiology, University College London, London, UK
| | - Paolo Denti
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Timothy D McHugh
- Centre for Clinical Microbiology, University College London, London, UK
| | - Frank Kloprogge
- Institute for Global Health, University College London, London, UK.
- Centre for Clinical Microbiology, University College London, London, UK.
| |
Collapse
|
9
|
Rigouts L, Condé M, Hassane-Harouna S, Bah K, Gumusboga M, Ruhwald M, Reenaers R, Fissette K, Gaichiri M, De Vos M, De Jong BC. Successful Mycobacterium tuberculosis culture isolation from tongue swabs: Results from both experimentally infected and clinical swabs from pulmonary tuberculosis patients. Diagn Microbiol Infect Dis 2024; 110:116423. [PMID: 39121811 DOI: 10.1016/j.diagmicrobio.2024.116423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/02/2024] [Accepted: 07/03/2024] [Indexed: 08/12/2024]
Abstract
This study explored Mycobacterium tuberculosis (MTB) growth from tongue swabs, both experimentally infected after sampling from healthy controls, or sampled from patients with smear-microscopy confirmed pulmonary tuberculosis (PTB). For both, we evaluated the performance of NALC-NaOH/MGIT960 (MGIT), Kudoh-Ogawa (KO), and cetylpyridinium chloride-Löwenstein-Jensen (CPC/LJ) culture processing methods. Experimentally spiked swabs from 20 participants exhibited 94.4% MTB growth when inoculated within 7 days of CPC exposure, declining significantly after 14-21 days (p<0.00001). KO-processed specimens showed 100% MTB growth, with a non-significant reduction after storage (94.1%; p=0.21), and all spiked swabs yielded growth in MGIT. In the field evaluation on 99 PTB patients, MGIT isolated MTB from 89% of tongue swabs, with an 8% contamination rate, compared to 99% MGIT positivity from sputum. Solid media had lower positivity, 62% for KO and 49% for CPC/LJ, suggesting MGIT as optimal for growing MTB from tongue swabs. Further testing of presumptive PTB patients is recommended.
Collapse
Affiliation(s)
- Leen Rigouts
- Mycobacteriology Unit, Institute of Tropical Medicine, Antwerp, Belgium; Biomedical Sciences Department, Antwerp University, Belgium.
| | - Moussa Condé
- Guinea National Reference Laboratory of Mycobacteriology, Conakry, Guinea
| | | | - Kindy Bah
- Guinea National Reference Laboratory of Mycobacteriology, Conakry, Guinea
| | - Mourad Gumusboga
- Mycobacteriology Unit, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Rabab Reenaers
- Mycobacteriology Unit, Institute of Tropical Medicine, Antwerp, Belgium
| | - Krista Fissette
- Mycobacteriology Unit, Institute of Tropical Medicine, Antwerp, Belgium
| | | | | | - Bouke C De Jong
- Mycobacteriology Unit, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
10
|
Gap-Gaupool B, Glenn SM, Milburn E, Turapov O, Crosatti M, Hincks J, Stewart B, Bacon J, Kendall SL, Voskuil MI, Riabova O, Monakhova N, Green J, Waddell SJ, Makarov VA, Mukamolova GV. Nitric oxide induces the distinct invisibility phenotype of Mycobacterium tuberculosis. Commun Biol 2024; 7:1206. [PMID: 39342050 PMCID: PMC11439070 DOI: 10.1038/s42003-024-06912-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
During infection Mycobacterium tuberculosis (Mtb) forms physiologically distinct subpopulations that are recalcitrant to treatment and undetectable using standard diagnostics. These difficult to culture or differentially culturable (DC) Mtb are revealed in liquid media, their revival is often stimulated by resuscitation-promoting factors (Rpf) and prevented by Rpf inhibitors. Here, we investigated the role of nitric oxide (NO) in promoting the DC phenotype. Rpf-dependent DC Mtb were detected following infection of interferon-γ-induced macrophages capable of producing NO, but not when inducible NO synthase was inactivated. After exposure of Mtb to a new donor for sustained NO release (named NOD), the majority of viable cells were Rpf-dependent and undetectable on solid media. Gene expression analyses revealed a broad transcriptional response to NOD, including down-regulation of all five rpf genes. The DC phenotype was partially reverted by over-expression of Rpfs which promoted peptidoglycan remodelling. Thus, NO plays a central role in the generation of Rpf-dependent Mtb, with implications for improving tuberculosis diagnostics and treatments.
Collapse
Affiliation(s)
- Brindha Gap-Gaupool
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Sarah M Glenn
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Emily Milburn
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Obolbek Turapov
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Marialuisa Crosatti
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Jennifer Hincks
- FACS Facility Core Biotechnology Services, University of Leicester, Leicester, LE1 9HN, UK
| | - Bradley Stewart
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK
| | - Joanna Bacon
- Discovery Group, Vaccine Development and Evaluation Centre, UK Health Security Agency, Porton Down, SP4 0JG, UK
| | - Sharon L Kendall
- Centre for Endemic, Emerging and Exotic Disease, the Royal Veterinary College, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Martin I Voskuil
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Olga Riabova
- Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia Monakhova
- Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Jeffrey Green
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Simon J Waddell
- Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, UK.
| | - Vadim A Makarov
- Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia.
| | - Galina V Mukamolova
- Leicester Tuberculosis Research Group, Department of Respiratory Sciences, University of Leicester, Leicester, LE1 9HN, UK.
- The National Institute for Health and Care Research Leicester Biomedical Research Centre, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
11
|
Mariner-Llicer C, Goig GA, Torres-Puente M, Vashakidze S, Villamayor LM, Saavedra-Cervera B, Mambuque E, Khurtsilava I, Avaliani Z, Rosenthal A, Gabrielian A, Shurgaia M, Shubladze N, García-Basteiro AL, López MG, Comas I. Genetic diversity within diagnostic sputum samples is mirrored in the culture of Mycobacterium tuberculosis across different settings. Nat Commun 2024; 15:7114. [PMID: 39237504 PMCID: PMC11377819 DOI: 10.1038/s41467-024-51266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Culturing and genomic sequencing of Mycobacterium tuberculosis (MTB) from tuberculosis (TB) cases is the basis for many research and clinical applications. The alternative, culture-free sequencing from diagnostic samples, is promising but poses challenges to obtain and analyse the MTB genome. Paradoxically, culture is assumed to impose a diversity bottleneck, which, if true, would entail unexplored consequences. To unravel this paradox we generate high-quality genomes of sputum-culture pairs from two different settings after developing a workflow for sequencing from sputum and a tailored bioinformatics analysis. Careful downstream comparisons reveal sources of sputum-culture incongruences due to false positive/negative variation associated with factors like low input MTB DNA or variable genomic depths. After accounting for these factors, contrary to the bottleneck dogma, we identify a 97% variant agreement within sputum-culture pairs, with a high correlation also in the variants' frequency (0.98). The combined analysis from five different settings and more than 100 available samples shows that our results can be extrapolated to different TB epidemic scenarios, demonstrating that for the cases tested culture accurately mirrors clinical samples.
Collapse
Affiliation(s)
| | - Galo A Goig
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
| | | | - Sergo Vashakidze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- The University of Georgia, Tbilisi, Georgia
| | - Luis M Villamayor
- FISABIO, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, València, Spain
| | - Belén Saavedra-Cervera
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Wellcome Sanger Institute, Hinxton, UK
| | - Edson Mambuque
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Iza Khurtsilava
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- European University, Tbilisi, Georgia
| | - Alex Rosenthal
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Andrei Gabrielian
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Marika Shurgaia
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Natalia Shubladze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- CIBERINFEC, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Barcelona, Spain
| | - Mariana G López
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
| | - Iñaki Comas
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
- CIBERESP, Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain.
| |
Collapse
|
12
|
Tarekegn BG, Tientcheu LD, Decker J, Bell AJ, Mukamolova GV, Kampmann B, Messele G, Abeje T, Aseffa A, Dockrell HM, Haldar P, Barer MR, Garton NJ. Host and pathogen factors that influence variability of Mycobacterium tuberculosis lipid body content in sputum from patients with tuberculosis: an observational study. THE LANCET. MICROBE 2024; 5:100885. [PMID: 38906163 DOI: 10.1016/s2666-5247(24)00108-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND High proportions of Mycobacterium tuberculosis cells in sputum containing triacylglycerol-rich lipid bodies have been shown to be associated with treatment failure or relapse following antituberculous chemotherapy. Although lipid body determination is a potential biomarker for supporting clinical trial and treatment decisions, factors influencing variability in sputum frequencies of lipid body-positive (%LB+) M tuberculosis in patients are unknown. We aimed to test our hypothesis that exposure to host-generated NO and M tuberculosis strains are factors associated with differences in sputum %LB+. METHODS In this observational study, we determined %LB+ frequencies before treatment by microscopy in patients with smear-positive tuberculosis from two separate prospective observational study settings (Gondar, Ethiopia, recruited between May 1, 2010, and April 30, 2011, and Fajara, The Gambia, who provided sputum samples before treatment between May 5, 2010, and Dec 22, 2011). In Ethiopia, fractional exhaled nitric oxide (FeNO) was measured as a biomarker of host NO, and M tuberculosis strain differences were determined by spoligotyping. Treatment response was assessed by percentage weight change after 7 months. In The Gambia, treatment responses were assessed as change in BMI and radiographic burden of disease after 6 months. Sputum M tuberculosis isolates were studied in vitro for their %LB+ and triacylglycerol synthase 1 (tgs1) mRNA responses to NO exposure. Propidium iodide staining was used as a measure of NO strain toxicity. Correlation between in vitro %LB+ frequencies following NO exposure and those of the same strain in sputum was examined with linear regression and Dunnett's multiple comparison test. FINDINGS In Ethiopia, 73 patients who were smear positive for pulmonary tuberculosis were recruited (43 [59%] were male and 30 [41%] were female). Of these, the %LB+ in the sputum of 59 patients showed linear correlation with log10 FeNO (r2=0·28; p<0·0001) and an association with strain spoligotype was suggested. Seven M tuberculosis strains from The Gambia showed different dose-responses to NO in vitro, demonstrated by changing lipid body content, tgs1 transcription, and bacterial toxicity. In sputum %LB+ frequencies correlated with in vitro %LB+ responses to NO of the corresponding isolate. In a subset of 34 patients across both cohorts, higher sputum %LB+ frequencies before treatment were associated with weaker responses to treatment than lower sputum %LB+ frequencies. INTERPRETATION M tuberculosis strain and exposure to host-generated NO are associated with sputum %LB+. Our results support the use of M tuberculosis strain-dependent sputum %LB+ as a predictive biomarker of treatment response. FUNDING The Medical Research Council, the University of Leicester, and the University of Gondar.
Collapse
Affiliation(s)
- Baye G Tarekegn
- Department of Respiratory Sciences, University of Leicester, Leicester, UK; Department of Medical Microbiology, University of Gondar, Gondar, Ethiopia
| | - Leopold D Tientcheu
- Medical Research Council Unit, The Gambia at London School of Hygiene & Tropical Medicine, Vaccines and Immunity Theme, Fajara, The Gambia; Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Jonathan Decker
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Andrew J Bell
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Galina V Mukamolova
- Department of Respiratory Sciences, University of Leicester, Leicester, UK; Leicester Tuberculosis Research Group, University of Leicester, Leicester, UK; National Institute for Health and Care Research Leicester Biomedical Research Centre, Leicester, UK
| | - Beate Kampmann
- Medical Research Council Unit, The Gambia at London School of Hygiene & Tropical Medicine, Vaccines and Immunity Theme, Fajara, The Gambia; Institut für Internationale Gesundheit and Centre for Global Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gashaw Messele
- Department of Surgery, University of Gondar, Gondar, Ethiopia
| | - Tadeye Abeje
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Hazel M Dockrell
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Pranabashis Haldar
- Department of Respiratory Sciences, University of Leicester, Leicester, UK; Leicester Tuberculosis Research Group, University of Leicester, Leicester, UK; National Institute for Health and Care Research Leicester Biomedical Research Centre, Leicester, UK
| | - Michael R Barer
- Department of Respiratory Sciences, University of Leicester, Leicester, UK; Leicester Tuberculosis Research Group, University of Leicester, Leicester, UK; National Institute for Health and Care Research Leicester Biomedical Research Centre, Leicester, UK; Department of Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Natalie J Garton
- Department of Respiratory Sciences, University of Leicester, Leicester, UK; Leicester Tuberculosis Research Group, University of Leicester, Leicester, UK; National Institute for Health and Care Research Leicester Biomedical Research Centre, Leicester, UK.
| |
Collapse
|
13
|
Gillespie SH, DiNardo AR, Georghiou SB, Sabiiti W, Kohli M, Panzner U, Kontsevaya I, Hittel N, Stuyver LJ, Tan JB, van Crevel R, Lange C, Thuong TNT, Heyckendorf J, Ruhwald M, Heinrich N. Developing biomarker assays to accelerate tuberculosis drug development: defining target product profiles. THE LANCET. MICROBE 2024; 5:100869. [PMID: 38735303 DOI: 10.1016/s2666-5247(24)00085-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 05/14/2024]
Abstract
Drug development for tuberculosis is hindered by the methodological limitations in the definitions of patient outcomes, particularly the slow organism growth and difficulty in obtaining suitable and representative samples throughout the treatment. We developed target product profiles for biomarker assays suitable for early-phase and late-phase clinical drug trials by consulting subject-matter experts on the desirable performance and operational characteristics of such assays for monitoring of tuberculosis treatment in drug trials. Minimal and optimal criteria were defined for scope, intended use, pricing, performance, and operational characteristics of the biomarkers. Early-stage trial assays should accurately quantify the number of viable bacilli, whereas late-stage trial assays should match the number, predict relapse-free cure, and replace culture conversion endpoints. The operational criteria reflect the infrastructure and resources available for drug trials. The effective tools should define the sterilising activity of the drug and lower the probability of treatment failure or relapse in people with tuberculosis. The target product profiles outlined in this Review should guide and de-risk the development of biomarker-based assays suitable for phase 2 and 3 clinical drug trials.
Collapse
Affiliation(s)
- Stephen H Gillespie
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK.
| | - Andrew R DiNardo
- Global TB Program, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, Netherlands
| | | | - Wilber Sabiiti
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, UK
| | | | - Ursula Panzner
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Irina Kontsevaya
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany; Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Norbert Hittel
- Janssen Global Public Health R&D, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | | | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, GA Nijmegen, Netherlands; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christoph Lange
- Global TB Program, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA; Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany; German Center for Infection Research (DZIF), Clinical Tuberculosis Unit, Borstel, Germany; Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany
| | | | - Jan Heyckendorf
- Respiratory Medicine & International Health, University of Lübeck, Lübeck, Germany; Clinic for Internal Medicine I, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | | | - Norbert Heinrich
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology, Infection and Pandemic Research, Munich, Germany
| |
Collapse
|
14
|
Wynn EA, Dide-Agossou C, Mubarak RA, 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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.607990. [PMID: 39229030 PMCID: PMC11370397 DOI: 10.1101/2024.08.20.607990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
A major challenge in tuberculosis (TB) therapeutics is that antibiotic exposure leads to changes in the physiologic state of M. tuberculosis (Mtb) which may enable the pathogen to withstand treatment. While antibiotic-treated Mtb have been evaluated in short-term in vitro experiments, it is unclear if and how long-term in vivo treatment with diverse antibiotics with varying treatment-shortening activity (sterilizing activity) affect Mtb physiologic states 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-week treatment with three sterilizing and three non-sterilizing antibiotics. Certain transcriptional changes were concordant 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 concordant 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 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 of cellular injury. In addition to elucidating 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, CO, USA
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
| | - Christian Dide-Agossou
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Reem Al Mubarak
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Karen Rossmassler
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Linda Crnic Institute for Down syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Victoria Ektnitphong
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Allison A Bauman
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lisa M Massoudi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Martin I Voskuil
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Gregory T Robertson
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Camille M Moore
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicholas D Walter
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- Consortium for Applied Microbial Metrics, Aurora, CO, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
15
|
Solanki P, Elton L, Honeyborne I, Park M, Satta G, McHugh TD. Improving the diagnosis of tuberculosis: old and new laboratory tools. Expert Rev Mol Diagn 2024; 24:487-496. [PMID: 38832527 DOI: 10.1080/14737159.2024.2362165] [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: 12/22/2023] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION Despite recent advances in diagnostic technologies and new drugs becoming available, tuberculosis (TB) remains a major global health burden. If detected early, screened for drug resistance, and fully treated, TB could be easily controlled. AREAS COVERED Here the authors discuss M. tuberculosis culture methods which are considered the definitive confirmation of M. tuberculosis infection, and limited advances made to build on these core elements of TB laboratory diagnosis. Literature searches showed that molecular techniques provide enhanced speed of turnaround, sensitivity, and richness of data. Sequencing of the whole genome, is becoming well established for identification and inference of drug resistance. PubMed® literature searches were conducted (November 2022-March 2024). EXPERT OPINION This section highlights future advances in diagnosis and infection control. Prevention of prolonged hospital admissions and rapid TAT are of the most benefit to the overall patient experience. Host transcriptional blood markers have been used in treatment monitoring studies and, with appropriate evaluation, could be rolled out in a diagnostic setting. Additionally, the MBLA is being incorporated into latest clinical trial designs. Whole genome sequencing has enhanced epidemiological evidence. Artificial intelligence, along with machine learning, have the ability to revolutionize TB diagnosis and susceptibility testing within the next decade.
Collapse
Affiliation(s)
- Priya Solanki
- UCL-TB and Centre for Clinical Microbiology, Division of Infection & Immunity, Royal Free Campus, London, UK
| | - Linzy Elton
- UCL-TB and Centre for Clinical Microbiology, Division of Infection & Immunity, Royal Free Campus, London, UK
| | - Isobella Honeyborne
- UCL-TB and Centre for Clinical Microbiology, Division of Infection & Immunity, Royal Free Campus, London, UK
| | - Mirae Park
- Respiratory Medicine, Imperial Healthcare NHS Trust, London, UK
| | - Giovanni Satta
- UCL-TB and Centre for Clinical Microbiology, Division of Infection & Immunity, Royal Free Campus, London, UK
| | - Timothy D McHugh
- UCL-TB and Centre for Clinical Microbiology, Division of Infection & Immunity, Royal Free Campus, London, UK
| |
Collapse
|
16
|
Xie YL, Eichberg C, Hapeela N, Nakabugo E, Anyango I, Arora K, Korte JE, Odero R, van Heerden J, Zemanay W, Kennedy S, Nabeta P, Hanif M, Rodrigues C, Skrahina A, Stevens W, Dietze R, Liu X, Ellner JJ, Alland D, Joloba ML, Schumacher SG, McCarthy KD, Nakiyingi L, Dorman SE. Xpert MTB/RIF Ultra versus mycobacterial growth indicator tube liquid culture for detection of Mycobacterium tuberculosis in symptomatic adults: a diagnostic accuracy study. THE LANCET. MICROBE 2024; 5:e520-e528. [PMID: 38608680 DOI: 10.1016/s2666-5247(24)00001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Xpert MTB/RIF Ultra (Ultra) is an automated molecular test for the detection of Mycobacterium tuberculosis in sputum. We compared the sensitivity of Ultra to that of mycobacterial growth indicator tube (MGIT) liquid culture, considered the most sensitive assay in routine clinical use. METHODS In this prospective, multicentre, cross-sectional diagnostic accuracy study, we used a non-inferiority design to assess whether the sensitivity of a single Ultra test was non-inferior to that of a single liquid culture for detection of M tuberculosis in sputum. We enrolled adults (age ≥18 years) with pulmonary tuberculosis symptoms in 11 countries and each adult provided three sputum specimens with a minimum volume of 2 mL over 2 days. Ultra was done directly on sputum 1, and Ultra and MGIT liquid culture were done on resuspended pellet from sputum 2. Results of MGIT and solid media cultures done on sputum 3 were considered the reference standard. The pre-defined non-inferiority margin was 5·0%. FINDINGS Between Feb 18, 2016, and Dec 4, 2019, we enrolled 2906 participants. 2600 (89%) participants were analysed, including 639 (25%) of 2600 who were positive for tuberculosis by the reference standard. Of the 2357 included in the non-inferiority analysis, 877 (37%) were HIV-positive and 984 (42%) were female. Sensitivity of Ultra performed directly on sputum 1 was non-inferior to that of sputum 2 MGIT culture (MGIT 91·1% vs Ultra 91·9%; difference -0·8 percentage points; 95% CI -2·8 to 1·1). Sensitivity of Ultra performed on sputum 2 pellet was also non-inferior to that of sputum 2 MGIT (MGIT 91·1% vs Ultra 91·9%; difference -0·8 percentage points; -2·7 to 1·0). INTERPRETATION For the detection of M tuberculosis in sputum from adults with respiratory symptoms, there was no difference in sensitivity of a single Ultra test to that of a single MGIT culture. Highly sensitive, rapid molecular approaches for M tuberculosis detection, combined with advances in genotypic methods for drug resistance detection, have potential to replace culture. FUNDING US National Institute of Allergy and Infectious Diseases.
Collapse
Affiliation(s)
- Yingda L Xie
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | | | - Nchimunya Hapeela
- Division of Medical Microbiology and Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Irene Anyango
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Kiranjot Arora
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | | | - Ronald Odero
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Judi van Heerden
- Division of Medical Microbiology and Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Widaad Zemanay
- Division of Medical Microbiology and Institute for Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Samuel Kennedy
- Medical University of South Carolina, Charleston, SC, USA
| | | | - Mahmud Hanif
- State TB Training and Demonstration Centre, New Delhi, India
| | | | - Alena Skrahina
- National Reference Laboratory, Republican Scientific and Practical Centre for Pulmonology and Tuberculosis, Minsk, Belarus
| | - Wendy Stevens
- Department of Molecular Medicine and Hematology, Faculty of Health Science, School of Pathology, and the National Priority Program of the National Health Laboratory Service, Johannesburg, South Africa
| | | | - Xin Liu
- Henan Provincial Chest Hospital, Zhengzhou, China
| | - Jerrold J Ellner
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - David Alland
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Moses L Joloba
- Mycobacteriology Laboratory, Department of Microbiology, School of Biomedical Sciences, Makerere University, Kampala, Uganda
| | | | | | - Lydia Nakiyingi
- Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Susan E Dorman
- Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
17
|
Espinosa-Pereiro J, Alagna R, Saluzzo F, González-Moreno J, Heinrich N, Sánchez-Montalvá A, Cirillo DM. A Systematic Review of Potential Biomarkers for Bacterial Burden and Treatment Efficacy Assessment in Tuberculosis Platform-Based Clinical Trials. J Infect Dis 2024; 229:1584-1595. [PMID: 37956107 DOI: 10.1093/infdis/jiad482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/28/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Adaptive platform trials can be more efficient than classic trials for developing new treatments. Moving from culture-based to simpler- or faster-to-measure biomarkers as efficacy surrogates may enhance this advantage. We performed a systematic review of treatment efficacy biomarkers in adults with tuberculosis. Platform trials can span different development phases. We grouped biomarkers as: α, bacterial load estimates used in phase 2a trials; β, early and end-of treatment end points, phase 2b-c trials; γ, posttreatment or trial-level estimates, phase 2c-3 trials. We considered as analysis unit (biomarker entry) each combination of biomarker, predicted outcome, and their respective measurement times or intervals. Performance metrics included: sensitivity, specificity, area under the receiver-operator curve (AUC), and correlation measures, and classified as poor, promising, or good. Eighty-six studies included 22 864 participants. From 1356 biomarker entries, 318 were reported with the performance metrics of interest, with 103 promising and 41 good predictors. Group results were: α, mycobacterial RNA and lipoarabinomannan (LAM) in sputum, and host metabolites in urine; β, mycobacterial RNA and host transcriptomic or cytokine signatures for early treatment response; and γ, host transcriptomics for recurrence. A combination of biomarkers from different categories could help in designing more efficient platform trials. Efforts to develop efficacy surrogates should be better coordinated.
Collapse
Affiliation(s)
- Juan Espinosa-Pereiro
- Infectious Diseases Department, Vall d'Hebrón University Hospital, Universitat Autónoma de Barcelona, Barcelona, Spain
- International Health Program, Catalan Institute of Health, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infeccioass, Instituto de Salud Carlos III, Madrid, Spain
| | - Riccardo Alagna
- San Raffaele Scientific Institute, Milan, Italy
- Qiagen, Srl, Milan, Italy
| | | | | | - Norbert Heinrich
- Center for International Health, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Infection Research, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig Maximilian University Munich (DZIF), Partner Site Munich, Munich, Germany
| | - Adrián Sánchez-Montalvá
- Infectious Diseases Department, Vall d'Hebrón University Hospital, Universitat Autónoma de Barcelona, Barcelona, Spain
- International Health Program, Catalan Institute of Health, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infeccioass, Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Estudio de Micobacterias, Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica, Madrid, Spain
| | | |
Collapse
|
18
|
Dartois V, Dick T. Therapeutic developments for tuberculosis and nontuberculous mycobacterial lung disease. Nat Rev Drug Discov 2024; 23:381-403. [PMID: 38418662 PMCID: PMC11078618 DOI: 10.1038/s41573-024-00897-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 03/02/2024]
Abstract
Tuberculosis (TB) drug discovery and development has undergone nothing short of a revolution over the past 20 years. Successful public-private partnerships and sustained funding have delivered a much-improved understanding of mycobacterial disease biology and pharmacology and a healthy pipeline that can tolerate inevitable attrition. Preclinical and clinical development has evolved from decade-old concepts to adaptive designs that permit rapid evaluation of regimens that might greatly shorten treatment duration over the next decade. But the past 20 years also saw the rise of a fatal and difficult-to-cure lung disease caused by nontuberculous mycobacteria (NTM), for which the drug development pipeline is nearly empty. Here, we discuss the similarities and differences between TB and NTM lung diseases, compare the preclinical and clinical advances, and identify major knowledge gaps and areas of cross-fertilization. We argue that applying paradigms and networks that have proved successful for TB, from basic research to clinical trials, will help to populate the pipeline and accelerate curative regimen development for NTM disease.
Collapse
Affiliation(s)
- Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA.
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, USA.
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
19
|
Gordhan BG, Padarath K, Sewcharran A, McIvor A, VanNieuwenhze MS, Waja Z, Martinson N, Kana BD. Clinical Strains of Mycobacterium tuberculosis Representing Different Genotype Families Exhibit Distinct Propensities to Adopt the Differentially Culturable State. Pathogens 2024; 13:318. [PMID: 38668273 PMCID: PMC11054447 DOI: 10.3390/pathogens13040318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/29/2024] Open
Abstract
Growing evidence points to the presence of differentially culturable tubercle bacteria (DCTB) in clinical specimens from individuals with active tuberculosis (TB) disease. These bacteria are unable to grow on solid media but can resuscitate in liquid media. Given the epidemiological success of certain clinical genotype families of Mycobacterium tuberculosis, we hypothesize that different strains may have distinct mechanisms of adaptation and tolerance. We used an in vitro carbon starvation model to determine the propensity of strains from lineages 2 and 4 that included the Beijing and LAM families respectively, to generate DCTB. Beijing strains were associated with a greater propensity to produce DCTB compared to LAM strains. Furthermore, LAM strains required culture filtrate (CF) for resuscitation whilst starved Beijing strains were not dependent on CF. Moreover, Beijing strains showed improved resuscitation with cognate CF, suggesting the presence of unique growth stimulatory molecules in this family. Analysis of starved Beijing and LAM strains showed longer cells, which with resuscitation were restored to a shorter length. Cell wall staining with fluorescent D-amino acids identified strain-specific incorporation patterns, indicating that cell surface remodeling during resuscitation was distinct between clinical strains. Collectively, our data demonstrate that M. tuberculosis clinical strains from different genotype lineages have differential propensities to generate DCTB, which may have implications for TB treatment success.
Collapse
Affiliation(s)
- Bhavna Gowan Gordhan
- Department of Science and Innovation and the National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2017, South Africa; (B.G.G.); (K.P.); (A.S.); (A.M.)
- National Health Laboratory Service, Johannesburg 2000, South Africa
| | - Kiyasha Padarath
- Department of Science and Innovation and the National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2017, South Africa; (B.G.G.); (K.P.); (A.S.); (A.M.)
- National Health Laboratory Service, Johannesburg 2000, South Africa
| | - Astika Sewcharran
- Department of Science and Innovation and the National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2017, South Africa; (B.G.G.); (K.P.); (A.S.); (A.M.)
- National Health Laboratory Service, Johannesburg 2000, South Africa
| | - Amanda McIvor
- Department of Science and Innovation and the National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2017, South Africa; (B.G.G.); (K.P.); (A.S.); (A.M.)
- National Health Laboratory Service, Johannesburg 2000, South Africa
| | | | - Ziyaad Waja
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg 2017, South Africa; (Z.W.); (N.M.)
| | - Neil Martinson
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg 2017, South Africa; (Z.W.); (N.M.)
- Center for TB Research, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bavesh Davandra Kana
- Department of Science and Innovation and the National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2017, South Africa; (B.G.G.); (K.P.); (A.S.); (A.M.)
- National Health Laboratory Service, Johannesburg 2000, South Africa
| |
Collapse
|
20
|
Patterson B, Dinkele R, Gessner S, Koch A, Hoosen Z, January V, Leonard B, McKerry A, Seldon R, Vazi A, Hermans S, Cobelens F, Warner DF, Wood R. Aerosolization of viable Mycobacterium tuberculosis bacilli by tuberculosis clinic attendees independent of sputum-Xpert Ultra status. Proc Natl Acad Sci U S A 2024; 121:e2314813121. [PMID: 38470917 PMCID: PMC10962937 DOI: 10.1073/pnas.2314813121] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/01/2024] [Indexed: 03/14/2024] Open
Abstract
Potential Mycobacterium tuberculosis (Mtb) transmission during different pulmonary tuberculosis (TB) disease states is poorly understood. We quantified viable aerosolized Mtb from TB clinic attendees following diagnosis and through six months' follow-up thereafter. Presumptive TB patients (n=102) were classified by laboratory, radiological, and clinical features into Group A: Sputum-Xpert Ultra-positive TB (n=52), Group B: Sputum-Xpert Ultra-negative TB (n=20), or Group C: TB undiagnosed (n=30). All groups were assessed for Mtb bioaerosol release at baseline, and subsequently at 2 wk, 2 mo, and 6 mo. Groups A and B were notified to the national TB program and received standard anti-TB chemotherapy; Mtb was isolated from 92% and 90% at presentation, 87% and 74% at 2 wk, 54% and 44% at 2 mo and 32% and 20% at 6 mo, respectively. Surprisingly, similar numbers were detected in Group C not initiating TB treatment: 93%, 70%, 48% and 22% at the same timepoints. A temporal association was observed between Mtb bioaerosol release and TB symptoms in all three groups. Persistence of Mtb bioaerosol positivity was observed in ~30% of participants irrespective of TB chemotherapy. Captured Mtb bacilli were predominantly acid-fast stain-negative and poorly culturable; however, three bioaerosol samples yielded sufficient biomass following culture for whole-genome sequencing, revealing two different Mtb lineages. Detection of viable aerosolized Mtb in clinic attendees, independent of TB diagnosis, suggests that unidentified Mtb transmitters might contribute a significant attributable proportion of community exposure. Additional longitudinal studies with sputum culture-positive and -negative control participants are required to investigate this possibility.
Collapse
Affiliation(s)
- Benjamin Patterson
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Ryan Dinkele
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Sophia Gessner
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Anastasia Koch
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Zeenat Hoosen
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Vanessa January
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Bryan Leonard
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Andrea McKerry
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Ronnett Seldon
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Andiswa Vazi
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| | - Sabine Hermans
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Frank Cobelens
- Amsterdam Institute for Global Health and Development, University of Amsterdam, Amsterdam1105, The Netherlands
| | - Digby F. Warner
- South African Medical Research Council, National Health Laboratory Service, University of Cape Town Molecular Mycobacteriology Research Unit & Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
| | - Robin Wood
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town7925, South Africa
- Aerobiology and TB Research Unit, Desmond Tutu Health Foundation, Cape Town7975, South Africa
| |
Collapse
|
21
|
Shenoy V, Gunda R, Noble C, Haraguchi A, Stevenson S, Daniel J. Fullertubes inhibit mycobacterial viability and prevent biofilm formation by disrupting the cell wall. Cell Biochem Funct 2024; 42:e3963. [PMID: 38424684 DOI: 10.1002/cbf.3963] [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: 09/21/2023] [Revised: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Mycobacterium tuberculosis and nontuberculous mycobacteria such as Mycobacterium abscessus cause diseases that are becoming increasingly difficult to treat due to emerging antibiotic resistance. The development of new antimicrobial molecules is vital for combating these pathogens. Carbon nanomaterials (CNMs) are a class of carbon-containing nanoparticles with promising antimicrobial effects. Fullertubes (C90 ) are novel carbon allotropes with a structure unique among CNMs. The effects of fullertubes on any living cell have not been studied. In this study, we demonstrate that pristine fullertube dispersions show antimicrobial effects on Mycobacterium smegmatis and M. abscessus. Using scanning electron microscopy, light microscopy, and molecular probes, we investigated the effects of these CNMs on mycobacterial cell viability, cellular integrity, and biofilm formation. C90 fullertubes at 1 µM inhibited mycobacterial viability by 97%. Scanning electron microscopy revealed that the cell wall structure of M. smegmatis and M. abscessus was severely damaged within 24 h of exposure to fullertubes. Additionally, exposure to fullertubes nearly abrogated the acid-fast staining property of M. smegmatis. Using SYTO-9 and propidium iodide, we show that exposure to the novel fullertubes compromises the integrity of the mycobacterial cell. We also show that the permeability of the mycobacterial cell wall was increased after exposure to fullertubes from our assays utilizing the molecular probe dichlorofluorescein and ethidium bromide transport. C90 fullertubes at 0.37 µM and C60 fullerenes at 0.56 µM inhibited pellicle biofilm formation by 70% and 90%, respectively. This is the first report on the antimycobacterial activities of fullertubes and fullerenes.
Collapse
Affiliation(s)
- Varun Shenoy
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
- Department of Chemistry and Biochemistry, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| | - Rashmika Gunda
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| | - Cora Noble
- Department of Chemistry and Biochemistry, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| | - Annalisa Haraguchi
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| | - Steven Stevenson
- Department of Chemistry and Biochemistry, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| | - Jaiyanth Daniel
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, Indiana, USA
| |
Collapse
|
22
|
Ferdosnejad K, Zamani MS, Soroush E, Fateh A, Siadat SD, Tarashi S. Tuberculosis and lung cancer: metabolic pathways play a key role. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:1262-1281. [PMID: 38305273 DOI: 10.1080/15257770.2024.2308522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/09/2024] [Accepted: 01/14/2024] [Indexed: 02/03/2024]
Abstract
Despite the fact that some cases of tuberculosis (TB) are undiagnosed and untreated, it remains a serious global public health issue. In the diagnosis, treatment, and control of latent and active TB, there may be a lack of effectiveness. An understanding of metabolic pathways can be fundamental to treat latent TB infection and active TB disease. Rather than targeting Mycobacterium tuberculosis, the control strategies aim to strengthen host responses to infection and reduce chronic inflammation by effectively enhancing host resistance to infection. The pathogenesis and progression of TB are linked to several metabolites and metabolic pathways, and they are potential targets for host-directed therapies. Additionally, metabolic pathways can contribute to the progression of lung cancer in patients with latent or active TB. A comprehensive metabolic pathway analysis is conducted to highlight lung cancer development in latent and active TB. The current study aimed to emphasize the association between metabolic pathways of tumor development in patients with latent and active TB. Health control programs around the world are compromised by TB and lung cancer due to their special epidemiological and clinical characteristics. Therefore, presenting the importance of lung cancer progression through metabolic pathways occurring upon TB infection can open new doors to improving control of TB infection and active TB disease while stressing that further evaluations are required to uncover this correlation.
Collapse
Affiliation(s)
| | | | - Erfan Soroush
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Samira Tarashi
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Ufimtseva EG, Eremeeva NI. Drug-Tolerant Mycobacterium tuberculosis Adopt Different Survival Strategies in Alveolar Macrophages of Patients with Pulmonary Tuberculosis. Int J Mol Sci 2023; 24:14942. [PMID: 37834390 PMCID: PMC10573496 DOI: 10.3390/ijms241914942] [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: 08/16/2023] [Revised: 08/31/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
The rapid spread of drug-resistant M. tuberculosis (Mtb) strains and the phenomenon of phenotypic tolerance to drugs present challenges toward achieving the goal of tuberculosis (TB) elimination worldwide. By using the ex vivo cultures of alveolar macrophages obtained from lung tissues of TB patients after intensive antimicrobial chemotherapy before surgery, different subpopulations of multidrug-tolerant Mtb with a spectrum of phenotypic and growth features were identified in the same TB lesions. Our results are indicative of not only passive mechanisms generating nonheritable resistance of Mtb to antibiotics, which are associated mainly with a lack of Mtb growth, but also some active mechanisms of Mtb persistence, such as cell wall and metabolic pathway remodeling. In one of the subpopulations, non-acid-fast Mtb have undergone significant reprogramming with the restoration of acid-fastness, lipoarabinomannan expression and replication in host cells of some patients after withdrawal of anti-TB drugs. Our data indicate the universal stress protein Rv2623 as a clinically relevant biomarker of Mtb that has lost acid-fastness in human lungs. The studies of Mtb survival, persistence, dormancy, and resumption and the identification of biomarkers characterizing these phenomena are very important concerning the development of vaccines and drug regimens with individualized management of patients for overcoming the resistance/tolerance crisis in anti-TB therapy.
Collapse
Affiliation(s)
- Elena G Ufimtseva
- Laboratory of Medical Biotechnology, Research Institute of Biochemistry, Federal Research Center of Fundamental and Translational Medicine, 2 Timakova Street, 630117 Novosibirsk, Russia
| | - Natalya I Eremeeva
- Institute of Disinfectology, F.F. Erisman Federal Scientific Center of Hygiene of the Federal Service on Surveillance for Consumer Rights Protection and Human Well-Being, 18a Nauchniy Proezd, 117246 Moscow, Russia
- Scientific Department, Ural Research Institute for Phthisiopulmonology, National Medical Research Center of Tuberculosis and Infectious Diseases of Ministry of Health of the Russian Federation, 50 XXII Partsyezda Street, 620039 Yekaterinburg, Russia
| |
Collapse
|
24
|
Wang P, Cai Y, Zhang G, Jiang L, Li Y. Establishment of an indirect ELISA for Mycobacterium tuberculosis MTB39A protein antibody. Appl Microbiol Biotechnol 2023; 107:6339-6349. [PMID: 37597020 PMCID: PMC10560182 DOI: 10.1007/s00253-023-12715-w] [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: 04/27/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/21/2023]
Abstract
The MTB39A protein is a member of the unique Mycobacterium tuberculosis (MTB) PE/PPE protein family and is the main candidate for tuberculosis (TB) diagnosis. The aim of this study was to establish a novel indirect ELISA (iELISA) method that uses antibodies against MTB. The MTB39A gene sequence was synthesized according to the MTB39A nucleotide sequence of the MTB H37Rv strain (GenBank accession number: NC_000962.3) and cloned into the pET28a( +) vector. After correct sequencing, it was transferred to Escherichia coli BL21 (DE3) receptor cells for expression and purification, and the purified recombinant protein was identified by SDS-PAGE and western blotting. The purified MTB39A protein was used as the capture antibody, and a rabbit polyclonal antibody against the MTB MTB39A protein was used as the detection antibody to establish an indirect ELISA method. The ELISA conditions were optimized, and the optimal coating concentration of the MTB39A antigen was determined to be 0.5 μg/mL. The optimal dilution of MTB39A rabbit polyclonal antibody was 1:4096, and the optimal dilution of HRP-goat anti-rabbit IgG was 1:4000. The results showed that this indirect ELISA method has high sensitivity, specificity and efficacy for MTB39A protein detection. Moreover, this indirect ELISA method has optimal stability and can be used for the initial detection of MTB antibodies in clinical human and bovine serum samples. The establishment of this assay provides a new method for the rapid diagnosis of MTB and technical support for the prevention and control of tuberculosis. KEY POINTS: • MTB MTB39A protein was expressed in a prokaryotic expression system. • Rabbit polyclonal antibody against MTB39A was prepared. • To establish an iELISA based on the MTB39A protein for the detection of MTB antibodies.
Collapse
Affiliation(s)
- Pu Wang
- School of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Yurong Cai
- School of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Gang Zhang
- School of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Lingling Jiang
- School of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China
| | - Yong Li
- School of Life Sciences, Ningxia University, Yinchuan, China.
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, China.
| |
Collapse
|
25
|
Ealand CS, Sewcharran A, Peters JS, Gordhan BG, Kamariza M, Bertozzi CR, Waja Z, Martinson NA, Kana BD. The performance of tongue swabs for detection of pulmonary tuberculosis. Front Cell Infect Microbiol 2023; 13:1186191. [PMID: 37743867 PMCID: PMC10512057 DOI: 10.3389/fcimb.2023.1186191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Oral and/or tongue swabs have demonstrated ability to detect Mycobacterium tuberculosis (Mtb) in adults with pulmonary tuberculosis (TB). Swabs provide useful alternative specimens for diagnosis of TB using molecular assays however, the diagnostic pickup by culture requires further improvement and development. Several studies identified the presence of differentially culturable tubercle bacilli (DCTB) populations in a variety of clinical specimens. These organisms do not grow in routine laboratory media and require growth factors in the form of culture filtrate (CF) from logarithmic phase cultures of Mtb H37Rv. Methods Herein, we compared the diagnostic performance of sputum and tongue swabs using Mycobacterial Growth Indicator Tube (MGIT) assays, Auramine smear, GeneXpert and DCTB assays supplemented with or without CF. Results From 89 eligible participants, 83 (93%), 66 (74%) and 79 (89%) were sputum positive by MGIT, smear and GeneXpert, respectively. The corresponding tongue swabs displayed a lower sensitivity with 39 (44%), 2 (2.0%) and 18 (20%) participants respectively for the same tests. We aimed to improve the diagnostic yield by utilizing DCTB assays. Sputum samples were associated with a higher positivity rate for CF-augmented DCTB at 82/89 (92%) relative to tongue swabs at 36/89 (40%). Similarly, sputum samples had a higher positivity rate for DCTB populations that were CF-independent at 64/89 (72%) relative to tongue swabs at 26/89 (29%). DCTB positivity increased significantly, relative to MGIT culture, for tongue swabs taken from HIV-positive participants. We next tested whether the use of an alternative smear stain, DMN-Trehalose, would improve diagnostic yield but noted no substantial increase. Discussion Collectively, our data show that while tongue swabs yield lower bacterial numbers for diagnostic testing, the use of growth supplementation may improve detection of TB particularly in HIV-positive people but this requires further interrogation in larger studies.
Collapse
Affiliation(s)
- Christopher S. Ealand
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Astika Sewcharran
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Julian S. Peters
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Bhavna G. Gordhan
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Mireille Kamariza
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Carolyn R. Bertozzi
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
- Department of Chemistry, Stanford University, Stanford, CA, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Ziyaad Waja
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
| | - Neil A. Martinson
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
- Perinatal HIV Research Unit (PHRU), University of the Witwatersrand, Johannesburg, South Africa
- Johns Hopkins University, Centre for Tuberculosis Research, Baltimore, MD, United States
| | - Bavesh D. Kana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| |
Collapse
|
26
|
Kim H, Choi HG, Shin SJ. Bridging the gaps to overcome major hurdles in the development of next-generation tuberculosis vaccines. Front Immunol 2023; 14:1193058. [PMID: 37638056 PMCID: PMC10451085 DOI: 10.3389/fimmu.2023.1193058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Although tuberculosis (TB) remains one of the leading causes of death from an infectious disease worldwide, the development of vaccines more effective than bacille Calmette-Guérin (BCG), the only licensed TB vaccine, has progressed slowly even in the context of the tremendous global impact of TB. Most vaccine candidates have been developed to strongly induce interferon-γ (IFN-γ)-producing T-helper type 1 (Th1) cell responses; however, accumulating evidence has suggested that other immune factors are required for optimal protection against Mycobacterium tuberculosis (Mtb) infection. In this review, we briefly describe the five hurdles that must be overcome to develop more effective TB vaccines, including those with various purposes and tested in recent promising clinical trials. In addition, we discuss the current knowledge gaps between preclinical experiments and clinical studies regarding peripheral versus tissue-specific immune responses, different underlying conditions of individuals, and newly emerging immune correlates of protection. Moreover, we propose how recently discovered TB risk or susceptibility factors can be better utilized as novel biomarkers for the evaluation of vaccine-induced protection to suggest more practical ways to develop advanced TB vaccines. Vaccines are the most effective tools for reducing mortality and morbidity from infectious diseases, and more advanced technologies and a greater understanding of host-pathogen interactions will provide feasibility and rationale for novel vaccine design and development.
Collapse
Affiliation(s)
- Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han-Gyu Choi
- Department of Microbiology and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
27
|
Lanni A, Iacobino A, Fattorini L, Giannoni F. Eradication of Drug-Tolerant Mycobacterium tuberculosis 2022: Where We Stand. Microorganisms 2023; 11:1511. [PMID: 37375013 PMCID: PMC10301435 DOI: 10.3390/microorganisms11061511] [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: 04/28/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The lungs of tuberculosis (TB) patients contain a spectrum of granulomatous lesions, ranging from solid and well-vascularized cellular granulomas to avascular caseous granulomas. In solid granulomas, current therapy kills actively replicating (AR) intracellular bacilli, while in low-vascularized caseous granulomas the low-oxygen tension stimulates aerobic and microaerophilic AR bacilli to transit into non-replicating (NR), drug-tolerant and extracellular stages. These stages, which do not have genetic mutations and are often referred to as persisters, are difficult to eradicate due to low drug penetration inside the caseum and mycobacterial cell walls. The sputum of TB patients also contains viable bacilli called differentially detectable (DD) cells that, unlike persisters, grow in liquid, but not in solid media. This review provides a comprehensive update on drug combinations killing in vitro AR and drug-tolerant bacilli (persisters and DD cells), and sterilizing Mycobacterium tuberculosis-infected BALB/c and caseum-forming C3HeB/FeJ mice. These observations have been important for testing new drug combinations in noninferiority clinical trials, in order to shorten the duration of current regimens against TB. In 2022, the World Health Organization, following the results of one of these trials, supported the use of a 4-month regimen for the treatment of drug-susceptible TB as a possible alternative to the current 6-month regimen.
Collapse
Affiliation(s)
| | | | | | - Federico Giannoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Via Regina Elena 299, 00161 Rome, Italy; (A.L.); (A.I.); (L.F.)
| |
Collapse
|
28
|
Chung ES, Kar P, Kamkaew M, Amir A, Aldridge BB. Mycobacterium tuberculosis grows linearly at the single-cell level with larger variability than model organisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541183. [PMID: 37292927 PMCID: PMC10245742 DOI: 10.1101/2023.05.17.541183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The ability of bacterial pathogens to regulate growth is crucial to control homeostasis, virulence, and drug response. Yet, we do not understand the growth and cell cycle behaviors of Mycobacterium tuberculosis (Mtb), a slow-growing pathogen, at the single-cell level. Here, we use time-lapse imaging and mathematical modeling to characterize these fundamental properties of Mtb. Whereas most organisms grow exponentially at the single-cell level, we find that Mtb exhibits a unique linear growth mode. Mtb growth characteristics are highly variable from cell-to-cell, notably in their growth speeds, cell cycle timing, and cell sizes. Together, our study demonstrates that growth behavior of Mtb diverges from what we have learned from model bacteria. Instead, Mtb generates a heterogeneous population while growing slowly and linearly. Our study provides a new level of detail into how Mtb grows and creates heterogeneity, and motivates more studies of growth behaviors in bacterial pathogens.
Collapse
|
29
|
Ayoun Alsoud R, Svensson RJ, Svensson EM, Gillespie SH, Boeree MJ, Diacon AH, Dawson R, Aarnoutse RE, Simonsson USH. Combined quantitative tuberculosis biomarker model for time-to-positivity and colony forming unit to support tuberculosis drug development. Front Pharmacol 2023; 14:1067295. [PMID: 36998606 PMCID: PMC10043246 DOI: 10.3389/fphar.2023.1067295] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/27/2023] [Indexed: 03/15/2023] Open
Abstract
Biomarkers are quantifiable characteristics of biological processes. In Mycobacterium tuberculosis, common biomarkers used in clinical drug development are colony forming unit (CFU) and time-to-positivity (TTP) from sputum samples. This analysis aimed to develop a combined quantitative tuberculosis biomarker model for CFU and TTP biomarkers for assessing drug efficacy in early bactericidal activity studies. Daily CFU and TTP observations in 83 previously patients with uncomplicated pulmonary tuberculosis after 7 days of different rifampicin monotherapy treatments (10–40 mg/kg) from the HIGHRIF1 study were included in this analysis. The combined quantitative tuberculosis biomarker model employed the Multistate Tuberculosis Pharmacometric model linked to a rifampicin pharmacokinetic model in order to determine drug exposure-response relationships on three bacterial sub-states using both the CFU and TTP data simultaneously. CFU was predicted from the MTP model and TTP was predicted through a time-to-event approach from the TTP model, which was linked to the MTP model through the transfer of all bacterial sub-states in the MTP model to a one bacterial TTP model. The non-linear CFU-TTP relationship over time was well predicted by the final model. The combined quantitative tuberculosis biomarker model provides an efficient approach for assessing drug efficacy informed by both CFU and TTP data in early bactericidal activity studies and to describe the relationship between CFU and TTP over time.
Collapse
Affiliation(s)
- Rami Ayoun Alsoud
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Robin J. Svensson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Elin M. Svensson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Stephen H. Gillespie
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Martin J. Boeree
- Department of Lung Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Rodney Dawson
- Division of Pulmonology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- University of Cape Town Lung Institute, Cape Town, South Africa
| | - Rob E. Aarnoutse
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ulrika S. H. Simonsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
- *Correspondence: Ulrika S. H. Simonsson,
| |
Collapse
|
30
|
Intranasal multivalent adenoviral-vectored vaccine protects against replicating and dormant M.tb in conventional and humanized mice. NPJ Vaccines 2023; 8:25. [PMID: 36823425 PMCID: PMC9948798 DOI: 10.1038/s41541-023-00623-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Viral-vectored vaccines are highly amenable for respiratory mucosal delivery as a means of inducing much-needed mucosal immunity at the point of pathogen entry. Unfortunately, current monovalent viral-vectored tuberculosis (TB) vaccine candidates have failed to demonstrate satisfactory clinical protective efficacy. As such, there is a need to develop next-generation viral-vectored TB vaccine strategies which incorporate both vaccine antigen design and delivery route. In this study, we have developed a trivalent chimpanzee adenoviral-vectored vaccine to provide protective immunity against pulmonary TB through targeting antigens linked to the three different growth phases (acute/chronic/dormancy) of Mycobacterium tuberculosis (M.tb) by expressing an acute replication-associated antigen, Ag85A, a chronically expressed virulence-associated antigen, TB10.4, and a dormancy/resuscitation-associated antigen, RpfB. Single-dose respiratory mucosal immunization with our trivalent vaccine induced robust, sustained tissue-resident multifunctional CD4+ and CD8+ T-cell responses within the lung tissues and airways, which were further quantitatively and qualitatively improved following boosting of subcutaneously BCG-primed hosts. Prophylactic and therapeutic immunization with this multivalent trivalent vaccine in conventional BALB/c mice provided significant protection against not only actively replicating M.tb bacilli but also dormant, non-replicating persisters. Importantly, when used as a booster, it also provided marked protection in the highly susceptible C3HeB/FeJ mice, and a single respiratory mucosal inoculation was capable of significant protection in a humanized mouse model. Our findings indicate the great potential of this next-generation TB vaccine strategy and support its further clinical development for both prophylactic and therapeutic applications.
Collapse
|
31
|
Peters JS, McIvor A, Papadopoulos AO, Masangana T, Gordhan BG, Waja Z, Otwombe K, Letutu M, Kamariza M, Sterling TR, Bertozzi CR, Martinson NA, Kana BD. Differentially culturable tubercle bacteria as a measure of tuberculosis treatment response. Front Cell Infect Microbiol 2023; 12:1064148. [PMID: 36710965 PMCID: PMC9877613 DOI: 10.3389/fcimb.2022.1064148] [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: 10/07/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Routine efficacy assessments of new tuberculosis (TB) treatments include quantitative solid culture or routine liquid culture, which likely miss quantification of drug tolerant bacteria. To improve these assessments, comparative analyses using additional measures such as quantification of differentially culturable tubercle bacteria (DCTB) are required. Essential for enabling this is a comparative measure of TB treatment responses using routine solid and liquid culture with liquid limiting dilutions (LLDs) that detect DCTB in sputum. Methods We recruited treatment-naïve TB patients, with and without HIV-infection, and serially quantified their sputum for DCTB over the course of treatment. Results Serial sputum sampling in 73 individuals during their first 14 days of treatment demonstrated that clearance of DCTB was slower compared to routine solid culture. Treatment response appeared to be characterized by four patterns: (1) Classic bi-phasic bacterial clearance; (2) early non-responders with slower clearance; (3) paradoxical worsening with an increase in bacterial count upon treatment initiation; and (4) non-responders with no change in bacterial load. During treatment, LLDs displayed greater bacterial yield when compared with quantitative solid culture. Upon treatment completion, 74% [46/62] of specimens displayed residual DCTB and within this group, two recurrences were diagnosed. Residual DCTB upon treatment completion was associated with a higher proportion of MGIT culture, GeneXpert, and smear positivity at two months post treatment. No recurrences occurred in the group without residual DCTB. Discussion These data indicate that DCTB assays detect distinct subpopulations of organisms in sputum that are missed by routine solid and liquid culture, and offer important alternatives for efficacy assessments of new TB treatments. The residual DCTB observed upon treatment completion suggests that TB treatment does not always eliminate all bacterial populations, a finding that should be investigated in larger cohorts.
Collapse
Affiliation(s)
- Julian S. Peters
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Amanda McIvor
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Andrea O. Papadopoulos
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Tshepiso Masangana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bhavna G. Gordhan
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ziyaad Waja
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa,School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Matebogo Letutu
- Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Mireille Kamariza
- Department of Biology, Stanford University, Stanford, CA, United States
| | | | - Carolyn R. Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, United States,Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Neil A. Martinson
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg, South Africa,Johns Hopkins University Center for TB Research, Baltimore, MD, United States
| | - Bavesh D. Kana
- Department of Science and Innovation/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, The National Health Laboratory Service, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,*Correspondence: Bavesh D. Kana,
| |
Collapse
|
32
|
Zainabadi K, Saito K, Mishra S, Walsh KF, Mathurin LD, Vilbrun SC, Ocheretina O, Pape JW, Fitzgerald DW, Nathan CF, Lee MH. Transcriptional Biomarkers of Differentially Detectable Mycobacterium tuberculosis in Patient Sputum. mBio 2022; 13:e0270122. [PMID: 36326252 PMCID: PMC9765512 DOI: 10.1128/mbio.02701-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Certain populations of Mycobacterium tuberculosis go undetected by standard diagnostics but can be enumerated using limiting dilution assays. These differentially detectable M. tuberculosis (DD M. tuberculosis) populations may have relevance for persistence due to their drug tolerance. It is unclear how well DD M. tuberculosis from patients is modeled by a recently developed in vitro model in which M. tuberculosis starved in phosphate-buffered saline is incubated with rifampin to produce DD M. tuberculosis (the PBS-RIF model). This study attempted to answer this question. We selected 14 genes that displayed differential expression in the PBS-RIF model and evaluated their expression in patient sputa containing various proportions of DD M. tuberculosis. The expression of 12/14 genes correlated with the relative abundance of DD M. tuberculosis in patient sputa. Culture filtrate (CF), which promotes recovery of DD M. tuberculosis from certain patient sputa, improved these correlations in most cases. The gene whose reduced expression relative to M. tuberculosis 16S rRNA showed the greatest association with the presence and relative abundance of DD M. tuberculosis in patient sputa, icl1, was recently shown to play a functional role in restraining DD M. tuberculosis formation in the PBS-RIF model. Expression of icl1, combined with two additional DD M. tuberculosis-related genes, showed strong performance for predicting the presence or absence of DD M. tuberculosis in patient sputa (receiver operating characteristic [ROC] area under the curve [AUC] = 0.88). Thus, the in vitro DD M. tuberculosis model developed by Saito et al. (K. Saito, T. Warrier, S. Somersan-Karakaya, L. Kaminski, et al., Proc Natl Acad Sci U S A 114:E4832-E4840, 2017, https://doi.org/10.1073/pnas.1705385114) bears a resemblance to DD M. tuberculosis found in tuberculosis (TB) patients, and DD M. tuberculosis transcriptional profiles may be useful for monitoring DD M. tuberculosis populations in patient sputum. IMPORTANCE Differentially detectable M. tuberculosis (DD M. tuberculosis), which is detectable by limiting dilution assays but not by CFU, is present and enriched for in TB patient sputum after initiation of first-line therapy. These cryptic cells may play a role in disease persistence due to their phenotypic tolerance to anti-TB drugs. A recently developed in vitro model of DD M. tuberculosis (the PBS-RIF model) has expanded our understanding of these cells, though how well it translates to DD M. tuberculosis in patients is currently unknown. To answer this question, we selected 14 genes that displayed differential expression in the PBS-RIF model and evaluated their expression in TB patient sputa. We found that 12/14 of these genes showed a similar expression profile in patient sputa that correlated with the relative abundance of DD M. tuberculosis. Further, the expression of three of these genes showed strong performance for predicting the presence or absence of DD M. tuberculosis in patient sputa. The use of DD M. tuberculosis transcriptional profiles may allow for easier monitoring of DD M. tuberculosis populations in patient sputum in comparison to limiting dilution assays.
Collapse
Affiliation(s)
- Kayvan Zainabadi
- Center for Global Health, Weill Cornell Medicine, New York, New York, USA
| | - Kohta Saito
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Saurabh Mishra
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
| | - Kathleen Frances Walsh
- Center for Global Health, Weill Cornell Medicine, New York, New York, USA
- Department of Medicine, Division of General Internal Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | | | - Oksana Ocheretina
- Center for Global Health, Weill Cornell Medicine, New York, New York, USA
| | - Jean William Pape
- Center for Global Health, Weill Cornell Medicine, New York, New York, USA
- Les Centres GHESKIO, Port-au-Prince, Haiti
| | | | - Carl F. Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, USA
| | - Myung Hee Lee
- Center for Global Health, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
33
|
Demina GR, Shleeva MO, Bagaeva DI, Vostroknutova GV, Kaprelyants AS. Detection of “Non-culturable” Mycobacterium tuberculosis Cells by Culture Methods. APPL BIOCHEM MICRO+ 2022. [DOI: 10.1134/s0003683822100064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
34
|
Mycobacterium tuberculosis Dormancy: How to Fight a Hidden Danger. Microorganisms 2022; 10:microorganisms10122334. [PMID: 36557586 PMCID: PMC9784227 DOI: 10.3390/microorganisms10122334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Both latent and active TB infections are caused by a heterogeneous population of mycobacteria, which includes actively replicating and dormant bacilli in different proportions. Dormancy substantially affects M. tuberculosis drug tolerance and TB clinical management due to a significant decrease in the metabolic activity of bacilli, which leads to the complexity of both the diagnosis and the eradication of bacilli. Most diagnostic approaches to latent infection deal with a subpopulation of active M. tuberculosis, underestimating the contribution of dormant bacilli and leading to limited success in the fight against latent TB. Moreover, active TB appears not only as a primary form of infection but can also develop from latent TB, when resuscitation from dormancy is followed by bacterial multiplication, leading to disease progression. To win against latent infection, the identification of the Achilles' heel of dormant M. tuberculosis is urgently needed. Regulatory mechanisms and metabolic adaptation to growth arrest should be studied using in vitro and in vivo models that adequately imitate latent TB infection in macroorganisms. Understanding the mechanisms underlying M. tuberculosis dormancy and resuscitation may provide clues to help control latent infection, reduce disease severity in patients, and prevent pathogen transmission in the population.
Collapse
|
35
|
Beltran CGG, Venter R, Mann TN, Davis JH, Kana BD, Walzl G. Culture filtrate supplementation can be used to improve Mycobacterium tuberculosis culture positivity for spinal tuberculosis diagnosis. Front Cell Infect Microbiol 2022; 12:1065893. [PMID: 36506008 PMCID: PMC9732374 DOI: 10.3389/fcimb.2022.1065893] [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: 10/10/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Culture remains the gold standard to diagnose spinal tuberculosis (STB) despite the paucibacillary nature of the disease. Current methods can take up to 42 days to yield a result, delaying the ability to rapidly detect drug resistance. Studies have demonstrated the use of supplementation with culture filtrate (CF) from an axenic culture of Mycobacterium tuberculosis (Mtb) as a source of growth factors to improve culture rates. Our objective was to test a modified culture assay, utilizing CF supplemented media (CFSM), to improve culture positivity rates for suspected STB. Twelve patients with suspected STB were assessed by conventional culture (BACTEC™ MGIT 960), GeneXpert™ and standard histopathological examination. Spinal biopsies were taken from areas of diseased vertebral tissue or abscess, predetermined from MRI. Additional biopsies were obtained to assess CFSM for improved detection and faster culture of Mtb. All cases were diagnosed as STB and treated empirically for tuberculosis based on either bacteriological evidence (GeneXpert™, MGIT and/or CFSM positive), or based on clinical presentation. 5 specimens (45.45%) were positive for Mtb DNA as detected by GeneXpert™ and 1 specimen (8.33%) was cultured using MGIT (time to detection; 18 days). CFSM was able to culture 7 specimens (58.3%), with all CFSM positive specimens yielding a culture within 14 days. Two samples were positive only using the CFSM assay pointing to additional yield for diagnostic workup. Modification of standard culture can improve detection of Mtb and reduce time to positivity in individuals with STB where culture material is a requirement.
Collapse
Affiliation(s)
- Caroline G. G. Beltran
- Department of Science and Technology-National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa,*Correspondence: Caroline G. G. Beltran,
| | - Rouxjeane Venter
- Department of Science and Technology-National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Theresa N. Mann
- Division of Orthopaedic Surgery, Department of Surgical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Johan H. Davis
- Division of Orthopaedic Surgery, Department of Surgical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Bavesh D. Kana
- Department of Science and Technology-National Research Foundation (DST-NRF) Centre of Excellence for Biomedical TB Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa,Medical Research Council Centre for the Aids Programme of Research in South Africa (MRC-CAPRISA) HIV-TB Pathogenesis and Treatment Research Unit, Centre for the AIDS Programme of Research in South Africa, CAPRISA, Durban, South Africa
| | - Gerhard Walzl
- Department of Science and Technology-National Research Foundation (DST-NRF) Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
36
|
Chengalroyen MD, Beukes GM, Otwombe K, Gordhan BG, Martinson N, Kana B. The detection of mixed tuberculosis infections using culture filtrate and resuscitation promoting factor deficient filtrate. Front Cell Infect Microbiol 2022; 12:1072073. [PMID: 36506007 PMCID: PMC9729742 DOI: 10.3389/fcimb.2022.1072073] [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: 10/17/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Tuberculosis (TB) infected individuals harbor a heterogenous population of differentially culturable tubercle bacilli (DCTB). Herein, we describe how DCTB assays using culture filtrate either containing or deficient in resuscitation promoting factors can uncover mixed infections. We demonstrate that Mycobacterium tuberculosis (Mtb) strain genotypes can be separated in DCTB assays based on their selective requirement for growth stimulatory factors. Beijing mixed infections appear to be associated with a higher bacterial load and reduced reliance on growth stimulatory factors. These data have important implications for identifying mixed infections and hetero-resistance, which in turn can affect selection of treatment regimen and establishment of transmission links.
Collapse
Affiliation(s)
- Melissa D. Chengalroyen
- National Health Laboratory Service, DST/NRF Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, Johannesburg, South Africa
| | - Germar M. Beukes
- National Health Laboratory Service, DST/NRF Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, Johannesburg, South Africa
| | - Kennedy Otwombe
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bhavna G. Gordhan
- National Health Laboratory Service, DST/NRF Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, Johannesburg, South Africa
| | - Neil Martinson
- National Health Laboratory Service, DST/NRF Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, Johannesburg, South Africa,Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa,Center for TB Research, Johns Hopkins University, Baltimore, MD, United States
| | - Bavesh Kana
- National Health Laboratory Service, DST/NRF Centre of Excellence for Biomedical TB Research, University of the Witwatersrand, Johannesburg, South Africa,*Correspondence: Bavesh Kana,
| |
Collapse
|
37
|
Mishra S, Saito K. Clinically encountered growth phenotypes of tuberculosis-causing bacilli and their in vitro study: A review. Front Cell Infect Microbiol 2022; 12:1029111. [PMID: 36439231 PMCID: PMC9684195 DOI: 10.3389/fcimb.2022.1029111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/20/2022] [Indexed: 07/11/2024] Open
Abstract
The clinical manifestations of tuberculosis (TB) vary widely in severity, site of infection, and outcomes of treatment-leading to simultaneous efforts to individualize therapy safely and to search for shorter regimens that can be successfully used across the clinical spectrum. In these endeavors, clinicians and researchers alike employ mycobacterial culture in rich media. However, even within the same patient, individual bacilli among the population can exhibit substantial variability in their culturability. Bacilli in vitro also demonstrate substantial heterogeneity in replication rate and cultivation requirements, as well as susceptibility to killing by antimicrobials. Understanding parallels in clinical, ex vivo and in vitro growth phenotype diversity may be key to identifying those phenotypes responsible for treatment failure, relapse, and the reactivation of bacilli that progresses TB infection to disease. This review briefly summarizes the current role of mycobacterial culture in the care of patients with TB and the ex vivo evidence of variability in TB culturability. We then discuss current advances in in vitro models that study heterogenous subpopulations within a genetically identical bulk culture, with an emphasis on the effect of oxidative stress on bacillary cultivation requirements. The review highlights the complexity that heterogeneity in mycobacterial growth brings to the interpretation of culture in clinical settings and research. It also underscores the intricacies present in the interplay between growth phenotypes and antimicrobial susceptibility. Better understanding of population dynamics and growth requirements over time and space promises to aid both the attempts to individualize TB treatment and to find uniformly effective therapies.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, United States
| | - Kohta Saito
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
38
|
Dartois VA, Rubin EJ. Anti-tuberculosis treatment strategies and drug development: challenges and priorities. Nat Rev Microbiol 2022; 20:685-701. [PMID: 35478222 PMCID: PMC9045034 DOI: 10.1038/s41579-022-00731-y] [Citation(s) in RCA: 197] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 12/12/2022]
Abstract
Despite two decades of intensified research to understand and cure tuberculosis disease, biological uncertainties remain and hamper progress. However, owing to collaborative initiatives including academia, the pharmaceutical industry and non-for-profit organizations, the drug candidate pipeline is promising. This exceptional success comes with the inherent challenge of prioritizing multidrug regimens for clinical trials and revamping trial designs to accelerate regimen development and capitalize on drug discovery breakthroughs. Most wanted are markers of progression from latent infection to active pulmonary disease, markers of drug response and predictors of relapse, in vitro tools to uncover synergies that translate clinically and animal models to reliably assess the treatment shortening potential of new regimens. In this Review, we highlight the benefits and challenges of 'one-size-fits-all' regimens and treatment duration versus individualized therapy based on disease severity and host and pathogen characteristics, considering scientific and operational perspectives.
Collapse
Affiliation(s)
- Véronique A Dartois
- Center for Discovery and Innovation, and Hackensack Meridian School of Medicine, Department of Medical Sciences, Hackensack Meridian Health, Nutley, NJ, USA.
| | - Eric J Rubin
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA
| |
Collapse
|
39
|
Mtafya B, Sabi I, John J, Sichone E, Olomi W, Gillespie SH, Ntinginya NE, Sabiiti W. Systematic assessment of clinical and bacteriological markers for tuberculosis reveals discordance and inaccuracy of symptom-based diagnosis for treatment response monitoring. Front Med (Lausanne) 2022; 9:992451. [DOI: 10.3389/fmed.2022.992451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/28/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundClinical symptoms are the benchmark of tuberculosis (TB) diagnosis and monitoring of treatment response but are not clear how they relate to TB bacteriology, particularly the novel tuberculosis-molecular bacterial load assay (TB-MBLA).MethodsPresumptive cases were bacteriologically confirmed for TB and assessed for symptoms and bacteriological resolution using smear microscopy (SM), culture, and TB-MBLA over 6-month treatment course. Kaplan–Meier and Kappa statistics were used to test the relationship between symptoms and bacteriological positivity.ResultsA cohort of 46 bacteriologically confirmed TB cases were analyzed for treatment response over a 6-month treatment course. Pre-treatment symptoms and bacteriological positivity concurred in over 70% of the cases. This agreement was lost in over 50% of cases whose chest pain, night sweat, and loss of appetite had resolved by week 2 of treatment. Cough resolved at a 3.2% rate weekly and was 0.3% slower than the combined bacteriological (average of MGIT and TB-MBLA positivity) resolution rate, 3.5% per week. A decrease in TB-MBLA positivity reflected a fall in bacillary load, 5.7 ± 1.3- at baseline to 0.30 ± 1.0- log10 eCFU/ml at month 6, and closer to cough resolution than other bacteriological measures, accounting for the only one bacteriologically positive case out of seven still coughing at month 6. Low baseline bacillary load patients were more likely to be bacteriologically negative, HR 5.6, p = 0.003 and HR 3.2, p = 0.014 by months 2 and 6 of treatment, respectively.ConclusionThe probability of clinical symptoms reflecting bacteriological positivity weakens as the patient progresses on anti-TB therapy, making the symptom-based diagnosis a less reliable marker of treatment response.
Collapse
|
40
|
Coleman M, Martinez L, Theron G, Wood R, Marais B. Mycobacterium tuberculosis Transmission in High-Incidence Settings-New Paradigms and Insights. Pathogens 2022; 11:1228. [PMID: 36364978 PMCID: PMC9695830 DOI: 10.3390/pathogens11111228] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 12/01/2023] Open
Abstract
Tuberculosis has affected humankind for thousands of years, but a deeper understanding of its cause and transmission only arose after Robert Koch discovered Mycobacterium tuberculosis in 1882. Valuable insight has been gained since, but the accumulation of knowledge has been frustratingly slow and incomplete for a pathogen that remains the number one infectious disease killer on the planet. Contrast that to the rapid progress that has been made in our understanding SARS-CoV-2 (the cause of COVID-19) aerobiology and transmission. In this Review, we discuss important historical and contemporary insights into M. tuberculosis transmission. Historical insights describing the principles of aerosol transmission, as well as relevant pathogen, host and environment factors are described. Furthermore, novel insights into asymptomatic and subclinical tuberculosis, and the potential role this may play in population-level transmission is discussed. Progress towards understanding the full spectrum of M. tuberculosis transmission in high-burden settings has been hampered by sub-optimal diagnostic tools, limited basic science exploration and inadequate study designs. We propose that, as a tuberculosis field, we must learn from and capitalize on the novel insights and methods that have been developed to investigate SARS-CoV-2 transmission to limit ongoing tuberculosis transmission, which sustains the global pandemic.
Collapse
Affiliation(s)
- Mikaela Coleman
- WHO Collaborating Centre for Tuberculosis and the Sydney Institute for Infectious Diseases, The University of Sydney, Sydney 2006, Australia
- Tuberculosis Research Program, Centenary Institute, The University of Sydney, Sydney 2050, Australia
| | - Leonardo Martinez
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| | - Grant Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7602, South Africa
| | - Robin Wood
- Desmond Tutu Health Foundation and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7700, South Africa
| | - Ben Marais
- WHO Collaborating Centre for Tuberculosis and the Sydney Institute for Infectious Diseases, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
41
|
Zainabadi K, Lee MH, Walsh KF, Vilbrun SC, Mathurin LD, Ocheretina O, Pape JW, Fitzgerald DW. An optimized method for purifying, detecting and quantifying Mycobacterium tuberculosis RNA from sputum for monitoring treatment response in TB patients. Sci Rep 2022; 12:17382. [PMID: 36253384 PMCID: PMC9574834 DOI: 10.1038/s41598-022-19985-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/07/2022] [Indexed: 02/05/2023] Open
Abstract
Diagnostics that more accurately detect and quantify viable Mycobacterium tuberculosis (Mtb) in the sputum of patients undergoing therapy are needed. Current culture- and molecular-based tests have shown limited efficacy for monitoring treatment response in TB patients, either due to the presence of viable sub-populations of Mtb which fail to grow under standard culture conditions (termed differentially detectable/culturable Mtb, DD Mtb) or the prolonged half-life of Mtb DNA in sputum. Here, we report an optimized RNA-based method for detecting and quantifying viable Mtb from patient sputum during the course of therapy. We first empirically derived a novel RNA extraction protocol from sputum that improves recovery of Mtb RNA while almost completely eliminating contamination from Mtb DNA and host nucleic acids. Next, we identified five Mtb 16S rRNA primer sets with varying limits of detection that were capable of distinguishing between live versus dead H37Rv Mtb. This combined protocol was then tested on sputa from a longitudinal cohort of patients receiving therapy for drug sensitive (DS) or drug resistant (DR) TB with first-line or second-line regimens, respectively. Results were compared with that of culture, including CFU, BACTEC MGIT, and a limiting dilution assay capable of detecting DD Mtb. The five 16S rRNA primer sets positively identified nearly all (range 94-100%) culture positive sputa, and a portion (19-37%) of culture negative sputa. In comparison, ten highly expressed Mtb mRNAs showed positivity in 72-86% of culture positive sputa, and in 0-13% of culture negative sputa. Two of the five 16S rRNA primer sets were able to positively identify 100% of culture positive sputa, and when tested on culture negative sputa from the DS cohort at 2 months post-initiation of therapy, identified 40% of samples as positive; a percentage that is in line with expected treatment failure rates when first-line therapy is discontinued early. These two primer sets also detected 16S rRNA in 13-20% of sputa at 6 months post-initiation of therapy in the DR cohort. Cycle threshold values for 16S rRNA showed a strong correlation with Mtb numbers as determined by culture (R > 0.87), including as Mtb numbers declined during the course of treatment with first-line and second-line regimens. The optimized molecular assay outlined here may have utility for monitoring treatment response in TB patients.
Collapse
Affiliation(s)
- Kayvan Zainabadi
- Center for Global Health, Weill Cornell Medicine, New York, NY, USA.
| | - Myung Hee Lee
- Center for Global Health, Weill Cornell Medicine, New York, NY, USA
| | - Kathleen Frances Walsh
- Center for Global Health, Weill Cornell Medicine, New York, NY, USA
- Division of General Internal Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | - Jean William Pape
- Center for Global Health, Weill Cornell Medicine, New York, NY, USA
- Les Centres GHESKIO, Port-au-Prince, Haiti
| | | |
Collapse
|
42
|
Akki M, Reddy DS, Katagi KS, Kumar A, Devarajegowda HC, M SK, Babagond V, Mane S, Joshi SD. Synthesis of coumarin-thioether conjugates as potential anti-tubercular agents: Their molecular docking and X-ray crystal studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
43
|
Liebenberg D, Gordhan BG, Kana BD. Drug resistant tuberculosis: Implications for transmission, diagnosis, and disease management. Front Cell Infect Microbiol 2022; 12:943545. [PMID: 36211964 PMCID: PMC9538507 DOI: 10.3389/fcimb.2022.943545] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/06/2022] [Indexed: 01/17/2023] Open
Abstract
Drug resistant tuberculosis contributes significantly to the global burden of antimicrobial resistance, often consuming a large proportion of the healthcare budget and associated resources in many endemic countries. The rapid emergence of resistance to newer tuberculosis therapies signals the need to ensure appropriate antibiotic stewardship, together with a concerted drive to develop new regimens that are active against currently circulating drug resistant strains. Herein, we highlight that the current burden of drug resistant tuberculosis is driven by a combination of ongoing transmission and the intra-patient evolution of resistance through several mechanisms. Global control of tuberculosis will require interventions that effectively address these and related aspects. Interrupting tuberculosis transmission is dependent on the availability of novel rapid diagnostics which provide accurate results, as near-patient as is possible, together with appropriate linkage to care. Contact tracing, longitudinal follow-up for symptoms and active mapping of social contacts are essential elements to curb further community-wide spread of drug resistant strains. Appropriate prophylaxis for contacts of drug resistant index cases is imperative to limit disease progression and subsequent transmission. Preventing the evolution of drug resistant strains will require the development of shorter regimens that rapidly eliminate all populations of mycobacteria, whilst concurrently limiting bacterial metabolic processes that drive drug tolerance, mutagenesis and the ultimate emergence of resistance. Drug discovery programs that specifically target bacterial genetic determinants associated with these processes will be paramount to tuberculosis eradication. In addition, the development of appropriate clinical endpoints that quantify drug tolerant organisms in sputum, such as differentially culturable/detectable tubercle bacteria is necessary to accurately assess the potential of new therapies to effectively shorten treatment duration. When combined, this holistic approach to addressing the critical problems associated with drug resistance will support delivery of quality care to patients suffering from tuberculosis and bolster efforts to eradicate this disease.
Collapse
|
44
|
Hammond RJH, Kloprogge F, Pasqua OD, Gillespie SH. Implications of drug-induced phenotypical resistance: Is isoniazid radicalizing M. tuberculosis? FRONTIERS IN ANTIBIOTICS 2022; 1:928365. [PMID: 39816411 PMCID: PMC11732046 DOI: 10.3389/frabi.2022.928365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2025]
Abstract
Background Tuberculosis treatment duration is long and does not guarantee eradication of infection. Shorter treatment regimens are a critical research objective to improve uptake and reduce the risk of relapse and bacterial resistance. The explanation for the need to continue treatment after patients are culture negative remains elusive. We have previously shown that the presence of lipid inclusions in mycobacterial cells is associated with an increase in antibiotic resistance. Aim We investigate the bactericidal effect of isoniazid and rifampicin on the expression of lipid inclusions and characterize the degree of the associated phenotypic antibiotic resistance to a range of anti-tuberculosis agents in current use. Methods Antibiotic killing effect for both M. tuberculosis and M. komossense were investigated by both hollow fiber bioreactor (HFS) studies and static time kill curve (STKC) experiments. Following STKC cultures were stained with resazurin, Sytox green and Nile red to establish their live/dead (resazurin positive/Sytox positive) and lipid inclusion status, respectively. In addition, M. komossense was studied in the hollow fiber bioreactor model (HFS) and exposed to isoniazid (H) and rifampicin (R). The MIC of current antituberculosis agents for cells from the treated hollow fiber experiments were tested. Results Antibiotic killing was similar for both species. For M. komossense; isoniazid was ineffective at the established MIC (1 mg/L) in the hollow fiber bioreactor but rifampicin reduced the viable count rapidly at MIC (0.4 mg/L). When the two drugs were combined at their respective MICs the killing effect was significant and greater than separately. Cells exposed to isoniazid (1x and 9x MIC) for 168 h showed considerable numbers of recoverable viable cells when compared with a combination of 1x MIC R & H where there were no viable cells detectable. For both drugs the number of lipid body positive cells increased over time and this effect was most pronounced for isoniazid and was associated with phenotypic resistance to multiple anti-tuberculosis drugs. Conclusion Our results showed that isoniazid is a potent stimulator of lipid body accumulation, culture persistence, and phenotypic resistance to multiple anti-tuberculosis drugs. These findings emphasize the importance of understanding mechanisms of drug-drug interactions and phenotypic resistance in regimen building.
Collapse
Affiliation(s)
- RJH Hammond
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Frank Kloprogge
- Clinical Pharmacology and Therapeutics Group, School of Pharmacy, University College London, London, United Kingdom
- Institute for Global Health, University College London, London, United Kingdom
| | - O. Della Pasqua
- Clinical Pharmacology and Therapeutics Group, School of Pharmacy, University College London, London, United Kingdom
| | - Stephen H. Gillespie
- Division of Infection and Global Health, School of Medicine, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
45
|
Gordhan BG, Sewcharran A, Letsoalo M, Chinappa T, Yende-Zuma N, Padayatchi N, Naidoo K, Kana BD. Detection of differentially culturable tubercle bacteria in sputum from drug-resistant tuberculosis patients. Front Cell Infect Microbiol 2022; 12:949370. [PMID: 36159642 PMCID: PMC9500503 DOI: 10.3389/fcimb.2022.949370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Several studies described the presence of non-replicating, drug-tolerant differentially culturable tubercle bacteria (DCTB) in sputum from patients with active tuberculosis (TB). These organisms are unable to form colonies on agar but can be recovered in liquid media supplemented with culture filtrate as a source of growth factors. Herein, we undertook to investigate the response of DCTB during the treatment of individuals with drug-resistant TB. A cohort of 100 participants diagnosed with rifampicin-resistant TB were enrolled and prospectively followed to monitor response to therapy using routine culture and limiting dilution assays, supplemented with culture filtrate (CF) to quantify DCTB. Fifteen participants were excluded due to contamination, and of the remaining 85 participants, 29, 49, and 7 were infected with rifampicin mono-resistant (RMR), multidrug-resistant (MDR), or extremely drug-resistant (XDR) TB, respectively. Analysis of baseline sputum demonstrated that CF supplementation of limiting dilution assays detected notable amounts of DCTB. Prevalence of DCTB was not influenced by smear status or mycobacterial growth indicator tube time to positivity. CF devoid of resuscitation promoting factors (Rpfs) yielded a greater amount of DCTB in sputum from participants with MDR-TB compared with those with RMR-TB. A similar effect was noted in DCTB assays without CF supplementation, suggesting that CF is dispensable for the detection of DCTB from drug-resistant strains. The HIV status of participants, and CD4 count, did not affect the amount of DCTB recovered. During treatment with second-line drug regimens, the probability of detecting DCTB from sputum specimens in liquid media with or without CF was higher compared with colony forming units, with DCTB detected up to 16 weeks post treatment. Collectively, these data point to differences in the ability of drug-resistant strains to respond to CF and Rpfs. Our findings demonstrate the possible utility of DCTB assays to diagnose and monitor treatment response for drug-resistant TB, particularly in immune compromised individuals with low CD4 counts.
Collapse
Affiliation(s)
- Bhavna G. Gordhan
- Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis (TB) Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Astika Sewcharran
- Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis (TB) Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
| | - Marothi Letsoalo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Thilgavathy Chinappa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Nonhlanhla Yende-Zuma
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- South African Medical Research Council (SAMRC)-Centre for the AIDS Programme of Research in South Africa (CAPRISA) Human Immunodeficiency Virus- Tuberculosis (HIV-TB) Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Nesri Padayatchi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- South African Medical Research Council (SAMRC)-Centre for the AIDS Programme of Research in South Africa (CAPRISA) Human Immunodeficiency Virus- Tuberculosis (HIV-TB) Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- South African Medical Research Council (SAMRC)-Centre for the AIDS Programme of Research in South Africa (CAPRISA) Human Immunodeficiency Virus- Tuberculosis (HIV-TB) Pathogenesis and Treatment Research Unit, University of KwaZulu-Natal, Durban, South Africa
| | - Bavesh D. Kana
- Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis (TB) Research, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and the National Health Laboratory Service, Johannesburg, South Africa
- *Correspondence: Bavesh D. Kana,
| |
Collapse
|
46
|
Alebouyeh S, Weinrick B, Achkar JM, García MJ, Prados-Rosales R. Feasibility of novel approaches to detect viable Mycobacterium tuberculosis within the spectrum of the tuberculosis disease. Front Med (Lausanne) 2022; 9:965359. [PMID: 36072954 PMCID: PMC9441758 DOI: 10.3389/fmed.2022.965359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB) is a global disease caused by Mycobacterium tuberculosis (Mtb) and is manifested as a continuum spectrum of infectious states. Both, the most common and clinically asymptomatic latent tuberculosis infection (LTBI), and the symptomatic disease, active tuberculosis (TB), are at opposite ends of the spectrum. Such binary classification is insufficient to describe the existing clinical heterogeneity, which includes incipient and subclinical TB. The absence of clinically TB-related symptoms and the extremely low bacterial burden are features shared by LTBI, incipient and subclinical TB states. In addition, diagnosis relies on cytokine release after antigenic T cell stimulation, yet several studies have shown that a high proportion of individuals with immunoreactivity never developed disease, suggesting that they were no longer infected. LTBI is estimated to affect to approximately one fourth of the human population and, according to WHO data, reactivation of LTBI is the main responsible of TB cases in developed countries. Assuming the drawbacks associated to the current diagnostic tests at this part of the disease spectrum, properly assessing individuals at real risk of developing TB is a major need. Further, it would help to efficiently design preventive treatment. This quest would be achievable if information about bacterial viability during human silent Mtb infection could be determined. Here, we have evaluated the feasibility of new approaches to detect viable bacilli across the full spectrum of TB disease. We focused on methods that specifically can measure host-independent parameters relying on the viability of Mtb either by its direct or indirect detection.
Collapse
Affiliation(s)
- Sogol Alebouyeh
- Department of Preventive Medicine and Public Health and Microbiology, Autonoma University of Madrid, Madrid, Spain
| | | | - Jacqueline M. Achkar
- Departments of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria J. García
- Department of Preventive Medicine and Public Health and Microbiology, Autonoma University of Madrid, Madrid, Spain
- *Correspondence: Maria J. García,
| | - Rafael Prados-Rosales
- Department of Preventive Medicine and Public Health and Microbiology, Autonoma University of Madrid, Madrid, Spain
- Rafael Prados-Rosales,
| |
Collapse
|
47
|
Paliwal D, Thom M, Hussein A, Ravishankar D, Wilkes A, Charleston B, Jones IM. Towards Reverse Vaccinology for Bovine TB: High Throughput Expression of Full Length Recombinant Mycobacterium bovis Proteins. Front Mol Biosci 2022; 9:889667. [PMID: 36032666 PMCID: PMC9402895 DOI: 10.3389/fmolb.2022.889667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis caused by Mycobacterium bovis, is a significant global pathogen causing economic loss in livestock and zoonotic TB in man. Several vaccine approaches are in development including reverse vaccinology which uses an unbiased approach to select open reading frames (ORF) of potential vaccine candidates, produce them as recombinant proteins and assesses their immunogenicity by direct immunization. To provide feasibility data for this approach we have cloned and expressed 123 ORFs from the M. bovis genome, using a mixture of E. coli and insect cell expression. We used a concatenated open reading frames design to reduce the number of clones required and single chain fusion proteins for protein pairs known to interact, such as the members of the PPE-PE family. Over 60% of clones showed soluble expression in one or the other host and most allowed rapid purification of the tagged bTB protein from the host cell background. The catalogue of recombinant proteins represents a resource that may be suitable for test immunisations in the development of an effective bTB vaccine.
Collapse
Affiliation(s)
- Deepa Paliwal
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Areej Hussein
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Alex Wilkes
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading, United Kingdom
- *Correspondence: Ian M. Jones,
| |
Collapse
|
48
|
Correlation between Sputum Bacterial Culture Positive Rate and Drug Sensitivity Test Results and Disease Severity inInpatients and Its Clinical Significance: A SystematicReview and Meta-Analysis. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:5102100. [PMID: 35875740 PMCID: PMC9303097 DOI: 10.1155/2022/5102100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022]
Abstract
Objective. To systematically evaluate the correlation between the positive rate of sputum bacterial culture and the results of drug sensitivity test and the severity of the disease and its clinical significance, so as to provide evidence-based medicine for clinical application. Methods. PubMed, Embase, ScienceDirect, Cochrane Library, China Knowledge Network Database (CNKI), China VIP Database, Wanfang Database, and China Biomedical Literature Database (CBM) online database were used. The retrieval time limit was from the establishment of the database to the present. Data for all included studies were extracted by two independent researchers, and the risk of bias for the quality of each included study was assessed by the Cochrane Handbook 5.1.0 criteria. RevMan5.4 statistical software was used to analyze the collected data by meta. Results. In the end, 6 RCT articles were included. Overall, 613 samples were included in 6 RCT studies. The correlation between the positive rate of sputum bacterial culture in inpatients and the severity of the disease was meta-analyzed. The heterogeneity test results showed that Chi2 = 177.20, df = 3, P < 0.00001, and I2 = 98%, indicating that there was obvious heterogeneity among the included research data. It was considered that there was a correlation between the positive rate of sputum bacterial culture and the severity of the disease. The correlation between the results of the drug sensitivity test of inpatients and the severity of the disease was evaluated. The results of the heterogeneity test showed that Chi2 = 0.00, df = 1, P = 1 > 0.05, and I2 = 0%, indicating that there was no heterogeneity among the included research data. In addition, the combined effect of WMD was analyzed by the fixed effect model. The combined effect dose WMD test was Z = 6.58 (P < 0.00001). It was considered that there was a correlation between the results of the drug sensitivity test and the severity of the disease. Conclusion. There is a correlation between positive sputum culture and drug sensitivity test results and the severity of the disease in hospitalized patients. In clinical practice, for hospitalized patients, the positive sputum bacterial culture rate and drug sensitivity test results can be used to guide the appropriate use of antibiotics. Due to the low input from the literature, more studies with higher methodological quality and longer follow-up are needed for further validation.
Collapse
|
49
|
Rodriguez GM, Sharma N, Biswas A, Sharma N. The Iron Response of Mycobacterium tuberculosis and Its Implications for Tuberculosis Pathogenesis and Novel Therapeutics. Front Cell Infect Microbiol 2022; 12:876667. [PMID: 35646739 PMCID: PMC9132128 DOI: 10.3389/fcimb.2022.876667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/25/2022] [Indexed: 12/16/2022] Open
Abstract
Most pathogenic bacteria require iron for growth. However, this metal is not freely available in the mammalian host. Due to its poor solubility and propensity to catalyze the generation of reactive oxygen species, host iron is kept in solution bound to specialized iron binding proteins. Access to iron is an important factor in the outcome of bacterial infections; iron limitation frequently induces virulence and drives pathogenic interactions with host cells. Here, we review the response of Mycobacterium tuberculosis to changes in iron availability, the relevance of this response to TB pathogenesis, and its potential for the design of new therapeutic interventions.
Collapse
|
50
|
Dide-Agossou C, Bauman AA, Ramey ME, Rossmassler K, Al Mubarak R, Pauly S, Voskuil MI, Garcia-Cremades M, Savic RM, Nahid P, Moore CM, Tasneen R, Nuermberger EL, Robertson GT, Walter ND. Combination of Mycobacterium tuberculosis RS Ratio and CFU Improves the Ability of Murine Efficacy Experiments to Distinguish between Drug Treatments. Antimicrob Agents Chemother 2022; 66:e0231021. [PMID: 35311519 PMCID: PMC9017352 DOI: 10.1128/aac.02310-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/17/2022] [Indexed: 11/20/2022] Open
Abstract
Murine tuberculosis drug efficacy studies have historically monitored bacterial burden based on CFU of Mycobacterium tuberculosis in lung homogenate. In an alternative approach, a recently described molecular pharmacodynamic marker called the RS ratio quantifies drug effect on a fundamental cellular process, ongoing rRNA synthesis. Here, we evaluated the ability of different pharmacodynamic markers to distinguish between treatments in three BALB/c mouse experiments at two institutions. We confirmed that different pharmacodynamic markers measure distinct biological responses. We found that a combination of pharmacodynamic markers distinguishes between treatments better than any single marker. The combination of the RS ratio with CFU showed the greatest ability to recapitulate the rank order of regimen treatment-shortening activity, providing proof of concept that simultaneous assessment of pharmacodynamic markers measuring different properties will enhance insight gained from animal models and accelerate development of new combination regimens. These results suggest potential for a new era in which antimicrobial therapies are evaluated not only on culture-based measures of bacterial burden but also on molecular assays that indicate how drugs impact the physiological state of the pathogen.
Collapse
Affiliation(s)
| | - Allison A. Bauman
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Michelle E. Ramey
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Karen Rossmassler
- Rocky Mountain Regional VA Medical Center, 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
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Samantha Pauly
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Martin I. Voskuil
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
| | - Maria Garcia-Cremades
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Rada M. Savic
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, California, USA
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, California, USA
| | - Payam Nahid
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, California, USA
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, California, USA
- UCSF Center for Tuberculosis, San Francisco, California, USA
| | - Camille M. Moore
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA
| | - Rokeya Tasneen
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Eric L. Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gregory T. Robertson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
| | - Nicholas D. Walter
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Consortium for Applied Microbial Metrics, Aurora, Colorado, USA
| |
Collapse
|