1
|
Liu Z, Hou J, Xie W, Deng H, Wang L. Regulation of sulfur starvation-induced proteins. 3 Biotech 2025; 15:130. [PMID: 40255445 PMCID: PMC12003235 DOI: 10.1007/s13205-025-04303-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
The application of environmentally friendly biological desulfurization (BDS) methods for treating organic sulfur offers significant potential to enhance the utilization of organic sulfur, reduce waste generation, and mitigate environmental harm. Several strains, including Escherichia coli, Rhodococcus erythropolis, and Mycobacterium tuberculosis, have been studied for their sulfur metabolic pathways and regulatory proteins involved in BDS. However, the current understanding of these regulatory mechanisms remains limited. This review outlines the role played by sulfur transport, acquisition and assimilation during sulfur metabolism and biosulfuration in response to sulfur starvation. In addition, we outline the involvement of sulfur starvation-inducible proteins in the L-methionine biosynthetic system. We hope to provide initial insights into the regulation of sulfur starvation-induced proteins. By exploring sulfur metabolism regulatory proteins, this review aims to offer valuable insights for the industrial application of biodesulfurization and unlock new possibilities for future research in this field.
Collapse
Affiliation(s)
- Zixin Liu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049 China
| | - Jie Hou
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049 China
| | - Wenhai Xie
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049 China
| | - Hongkuan Deng
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049 China
| | - Lijuan Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049 China
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014 China
| |
Collapse
|
2
|
Dinh P, Tran C, Dinh T, Ha HA, Utegenova A, Ali A, Alamri A. Identification and assessment of hub genes and miRNAs coregulatory associated with immune infiltrations and drug interactions in latent tuberculosis based on MicroarrayData analysis, molecular docking, and dynamic simulation. Biochem Biophys Rep 2025; 41:101952. [PMID: 40034257 PMCID: PMC11875834 DOI: 10.1016/j.bbrep.2025.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/01/2025] [Accepted: 02/09/2025] [Indexed: 03/05/2025] Open
Abstract
Tuberculosis (TB) remains a major global health concern, with the transition from latent to active TB still poorly understood. Therefore, enhancing clinical management and prevention strategies for TB is essential. High-throughput sequencing data of genes and miRNAs from individuals at different TB stages were obtained from NCBI. Differential expression analysis was performed using the R package limma, alongside GO and KEGG analyses. The central regulatory network of miRNAs was visualized with Cytoscape, and relevant genes were validated using ROC analysis. The predicted key genes involved in the transition from latent to active TB, including PLEKHG1, CLPB, DOK4, IL1β, and TLR3, are primarily associated with multicellular organism processes, stimulus-response, GPCR ligand binding, and immune functions. Finally, we screened Celastrol and Cefaclor Anhydrous targeting IL1β as potent anti-inflammatory drug to reduce the inflammation due to TB. These findings were further validated with Molecular dynamic simulation MM-GBSA and PCA analysis. Our study advances the understanding of latent tuberculosis and identifies genes and microRNAs as potential biomarkers for diagnosis, monitoring, and treatment, with broader implications for complex disease research.
Collapse
Affiliation(s)
- PhongSon Dinh
- College of Medicine and Pharmacy, Duy Tan University, Danang, 550000, Viet Nam
| | - ChauMyThanh Tran
- College of Medicine and Pharmacy, Duy Tan University, Danang, 550000, Viet Nam
| | - ThiPhuongHoai Dinh
- Department of Neurosurgery, Hue University Hospital, Hue University of Medicine and Pharmacy, Hue University, Hue, 530000, Viet Nam
| | - Hai-Anh Ha
- College of Medicine and Pharmacy, Duy Tan University, Danang, 550000, Viet Nam
| | - Aigul Utegenova
- Department of Microbiology and Virology, Astana Medical University, Astana, 010000, Kazakhstan
| | - Awais Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 2300, Pakistan
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
3
|
Singh DK, Ahmed M, Akter S, Shivanna V, Bucşan AN, Mishra A, Golden NA, Didier PJ, Doyle LA, Hall-Ursone S, Roy CJ, Arora G, Dick EJ, Jagannath C, Mehra S, Khader SA, Kaushal D. Prevention of tuberculosis in cynomolgus macaques by an attenuated Mycobacterium tuberculosis vaccine candidate. Nat Commun 2025; 16:1957. [PMID: 40000643 PMCID: PMC11861635 DOI: 10.1038/s41467-025-57090-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The need for novel vaccination strategies to control tuberculosis (TB) is underscored by the limited and variable efficacy of the currently licensed vaccine, Bacille Calmette-Guerin (BCG). SigH is critical for Mycobacterium tuberculosis (Mtb) to mitigate oxidative stress, and in its absence Mtb is unable to scavenge host oxidative/nitrosative bursts. The MtbΔsigH (ΔsigH) isogenic mutant induces signatures of the innate immunity in macrophages and protects rhesus macaques from a lethal Mtb challenge. To understand the immune mechanisms of protection via mucosal vaccination with ΔsigH, we employed the resistant cynomolgus macaque model; and our results show that ΔsigH vaccination significantly protects against lethal Mtb challenge in this species. ΔsigH-vaccinated macaques are devoid of granulomas and instead generate inducible bronchus associated lymphoid structures, and robust antigen-specific CD4+ and CD8+ T cell responses, driven by a hyper-immune, trained immunity-like phenotype in host macrophages with enhanced antigen presentation. Correlates of protection in ΔsigH-vaccinated macaques include gene signatures of T cell activation, IFNG production, including IFN-responsive, activated T cells, concomitant with IFNG production, and suppression of IDO+ Type I IFN-responsive macrophage recruitment. Thus, ΔsigH is a promising lead candidate for further development as an antitubercular vaccine.
Collapse
Affiliation(s)
- Dhiraj K Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Mushtaq Ahmed
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Sadia Akter
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Vinay Shivanna
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Allison N Bucşan
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Nadia A Golden
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Peter J Didier
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Lara A Doyle
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Shannan Hall-Ursone
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chad J Roy
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Garima Arora
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, USA
| | - Shabaana A Khader
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA.
| |
Collapse
|
4
|
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
|
5
|
Zhang Q, Zhu W, He S, Lei J, Xu L, Hu S, Zhang Z, Cai D, Chen S. Understanding energy fluctuation during the transition state: The role of AbrB in Bacillus licheniformis. Microb Cell Fact 2024; 23:296. [PMID: 39491006 PMCID: PMC11533420 DOI: 10.1186/s12934-024-02572-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Limited research has been conducted on energy fluctuation during the transition state, despite the critical role of energy supply in microbial physiological metabolism. RESULTS This study aimed to investigate the regulatory function of transition state transcription factor AbrB on energy metabolism in Bacillus licheniformis WX-02. Firstly, the deletion of abrB was found to prolong the cell generation time, significantly reducing the intercellular ATP concentration and NADH/NAD+ ratio at the early stage. Subsequently, various target genes and transcription factors regulated by AbrB were identified through in vitro verification assays. Specifically, AbrB was shown to modulate energy metabolism by directly regulating the expression of genes pyk and pgk in substrate-level phosphorylation, as well as genes narK and narGHIJ associated with nitrate respiration. In terms of oxidative phosphorylation, AbrB not only directly regulated ATP generation genes, including cyd, atpB, hmp, ndh, qoxA and sdhC, but also influenced the expression of NAD-dependent enzymes and intracellular NADH/NAD+ ratio. Additionally, AbrB positively affected the expression of transcription factors CcpN, Fnr, Rex, and ResD involved in energy supply, while negatively affected the regulator CcpA. Overall, this study found that AbrB positively regulates both substrate-level phosphorylation and oxidative phosphorylation, while negatively regulating nitrate respiration. CONCLUSIONS This study proposes a comprehensive regulatory network of AbrB on energy metabolism in Bacillus, expanding the understanding of regulatory mechanisms of AbrB and elucidating energy fluctuations during the transition state.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Wanying Zhu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Shisi He
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Jiaqi Lei
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Liangsheng Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Shiying Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Zheng Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China
| | - Dongbo Cai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China.
| | - Shouwen Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Environmental Microbial Technology Center of Hubei Province, College of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan, Hubei, 430062, PR China.
| |
Collapse
|
6
|
Bustad E, Petry E, Gu O, Griebel BT, Rustad TR, Sherman DR, Yang JH, Ma S. Predicting bacterial fitness in Mycobacterium tuberculosis with transcriptional regulatory network-informed interpretable machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614645. [PMID: 39386570 PMCID: PMC11463588 DOI: 10.1101/2024.09.23.614645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis disease, the greatest source of global mortality by a bacterial pathogen. Mtb adapts and responds to diverse stresses such as antibiotics by inducing transcriptional stress-response regulatory programs. Understanding how and when these mycobacterial regulatory programs are activated could enable novel treatment strategies for potentiating the efficacy of new and existing drugs. Here we sought to define and analyze Mtb regulatory programs that modulate bacterial fitness. We assembled a large Mtb RNA expression compendium and applied these to infer a comprehensive Mtb transcriptional regulatory network and compute condition-specific transcription factor activity profiles. We utilized transcriptomic and functional genomics data to train an interpretable machine learning model that can predict Mtb fitness from transcription factor activity profiles. We demonstrated that this transcription factor activity-based model can successfully predict Mtb growth arrest and growth resumption under hypoxia and reaeration using only RNA-seq expression data as a starting point. These integrative network modeling and machine learning analyses thus enable the prediction of mycobacterial fitness under different environmental and genetic contexts. We envision these models can potentially inform the future design of prognostic assays and therapeutic intervention that can cripple Mtb growth and survival to cure tuberculosis disease.
Collapse
Affiliation(s)
- Ethan Bustad
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| | - Edson Petry
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Oliver Gu
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Braden T. Griebel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
| | | | - David R. Sherman
- Department of Microbiology, University of Washington, Seattle WA, USA
| | - Jason H. Yang
- Center for Emerging and Re-emerging Pathogens, Rutgers New Jersey Medical School, Newark NJ, USA
- Department of Microbiology, Biochemistry, & Molecular Genetics, Rutgers New Jersey Medical School, Newark NJ, USA
| | - Shuyi Ma
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
- Department of Chemical Engineering, University of Washington, Seattle WA, USA
- Department of Pediatrics, University of Washington, Seattle WA, USA
- Pathobiology Graduate Program, Department of Global Health, University of Washington, Seattle WA, USA
| |
Collapse
|
7
|
Priyanka, Sharma S, Sharma M. Role of PE/PPE proteins of Mycobacterium tuberculosis in triad of host mitochondria, oxidative stress and cell death. Microb Pathog 2024; 193:106757. [PMID: 38908454 DOI: 10.1016/j.micpath.2024.106757] [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: 03/11/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The PE and PPE family proteins of Mycobacterium tuberculosis (Mtb) is exclusively found in pathogenic Mycobacterium species, comprising approximately 8-10 % of the Mtb genome. These emerging virulent factors have been observed to play pivotal roles in Mtb pathogenesis and immune evasion through various strategies. These immunogenic proteins are known to modulate the host immune response and cell-death pathways by targeting the powerhouse of the cell, the mitochondria to support Mtb survival. In this article, we are focused on how PE/PPE family proteins target host mitochondria to induce mitochondrial perturbations, modulate the levels of cellular ROS (Reactive oxygen species) and control cell death pathways. We observed that the time of expression of these proteins at different stages of infection is crucial for elucidating their impact on the cell death pathways and eventually on the outcome of infection. This article focuses on understanding the contributions of the PE/PPE proteins by unravelling the triad of host mitochondria, oxidative stress and cell death pathways that facilitate the Mtb persistence. Understanding the role of these proteins in host cellular pathways and the intricate mechanisms paves the way for the development of novel therapeutic strategies to combat TB infections.
Collapse
Affiliation(s)
- Priyanka
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Sadhna Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| | - Monika Sharma
- DSKC BioDiscovery Laboratory, Miranda House, and Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
8
|
Anjou C, Lotoux A, Morvan C, Martin-Verstraete I. From ubiquity to specificity: The diverse functions of bacterial thioredoxin systems. Environ Microbiol 2024; 26:e16668. [PMID: 38899743 DOI: 10.1111/1462-2920.16668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The thioredoxin (Trx) system, found universally, is responsible for the regeneration of reversibly oxidized protein thiols in living cells. This system is made up of a Trx and a Trx reductase, and it plays a central role in maintaining thiol-based redox homeostasis by reducing oxidized protein thiols, such as disulfide bonds in proteins. Some Trxs also possess a chaperone function that is independent of thiol-disulfide exchange, in addition to their thiol-disulfide reductase activity. These two activities of the Trx system are involved in numerous physiological processes in bacteria. This review describes the diverse physiological roles of the Trx system that have emerged throughout bacterial evolution. The Trx system is essential for responding to oxidative and nitrosative stress. Beyond this primary function, the Trx system also participates in redox regulation and signal transduction, and in controlling metabolism, motility, biofilm formation, and virulence. This range of functions has evolved alongside the diversity of bacterial lifestyles and their specific constraints. This evolution can be characterized by the multiplication of the systems and by the specialization of cofactors or targets to adapt to the constraints of atypical lifestyles, such as photosynthesis, insect endosymbiosis, or spore-forming bacteria.
Collapse
Affiliation(s)
- Cyril Anjou
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Aurélie Lotoux
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Claire Morvan
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
9
|
Sao Emani C, Reiling N. The efflux pumps Rv1877 and Rv0191 play differential roles in the protection of Mycobacterium tuberculosis against chemical stress. Front Microbiol 2024; 15:1359188. [PMID: 38516013 PMCID: PMC10956863 DOI: 10.3389/fmicb.2024.1359188] [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: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Background It was previously shown that GlnA3sc enabled Streptomyces coelicolor to survive in excess polyamines. However, subsequent studies revealed that Rv1878, the corresponding Mycobacterium tuberculosis (M.tb) ortholog, was not essential for the detoxification of spermine (Spm), in M.tb. On the other hand, the multi-drug efflux pump Rv1877 was previously shown to enable export of a wide range of compounds, while Rv0191 was shown to be more specific to chloramphenicol. Rationale Therefore, we first wanted to determine if detoxification of Spm by efflux can be achieved by any efflux pump, or if that was dependent upon the function of the pump. Next, since Rv1878 was found not to be essential for the detoxification of Spm, we sought to follow-up on the investigation of the physiological role of Rv1878 along with Rv1877 and Rv0191. Approach To evaluate the specificity of efflux pumps in the mycobacterial tolerance to Spm, we generated unmarked ∆rv1877 and ∆rv0191 M.tb mutants and evaluated their susceptibility to Spm. To follow up on the investigation of any other physiological roles they may have, we characterized them along with the ∆rv1878 M.tb mutant. Results The ∆rv1877 mutant was sensitive to Spm stress, while the ∆rv0191 mutant was not. On the other hand, the ∆rv1878 mutant grew better than the wild-type during iron starvation yet was sensitive to cell wall stress. The proteins Rv1877 and Rv1878 seemed to play physiological roles during hypoxia and acidic stress. Lastly, the ∆rv0191 mutant was the only mutant that was sensitive to oxidative stress. Conclusion The multidrug MFS-type efflux pump Rv1877 is required for Spm detoxification, as opposed to Rv0191 which seems to play a more specific role. Moreover, Rv1878 seems to play a role in the regulation of iron homeostasis and the reconstitution of the cell wall of M.tb. On the other hand, the sensitivity of the ∆rv0191 mutant to oxidative stress, suggests that Rv0191 may be responsible for the transport of low molecular weight thiols.
Collapse
Affiliation(s)
- Carine Sao Emani
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Norbert Reiling
- Microbial Interface Biology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
| |
Collapse
|
10
|
Cui Y, Dang G, Wang H, Tang Y, Lv M, Liu S, Song N. DosR's multifaceted role on Mycobacterium bovis BCG revealed through multi-omics. Front Cell Infect Microbiol 2023; 13:1292864. [PMID: 38076461 PMCID: PMC10703047 DOI: 10.3389/fcimb.2023.1292864] [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: 09/12/2023] [Accepted: 10/31/2023] [Indexed: 12/18/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular bacterium that causes a highly contagious and potentially lethal tuberculosis (TB) in humans. It can maintain a dormant TB infection within the host. DosR (dormancy survival regulator) (Rv3133c) has been recognized as one of the key transcriptional proteins regulating bacterial dormancy and participating in various metabolic processes. In this study, we extensively investigate the still not well-comprehended role and mechanism of DosR in Mycobacterium bovis (M. bovis) Bacillus Calmette-Guérin (BCG) through a combined omics analysis. Our study finds that deleting DosR significantly affects the transcriptional levels of 104 genes and 179 proteins. Targeted metabolomics data for amino acids indicate that DosR knockout significantly upregulates L-Aspartic acid and serine synthesis, while downregulating seven other amino acids, including L-histidine and lysine. This suggests that DosR regulates amino acid synthesis and metabolism. Taken together, these findings provide molecular and metabolic bases for DosR effects, suggesting that DosR may be a novel regulatory target.
Collapse
Affiliation(s)
- Yingying Cui
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Guanghui Dang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hui Wang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yiyi Tang
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Mingyue Lv
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Siguo Liu
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ningning Song
- State Key Laboratory for Animal Disease Control and Prevention, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Weifang Key Laboratory of Respiratory Tract Pathogens and Drug Therapy, Weifang, China
| |
Collapse
|
11
|
Fang J, Dong C, Xiong S. Mycobacterium tuberculosis Rv0790c inhibits the cellular autophagy at its early stage and facilitates mycobacterial survival. Front Cell Infect Microbiol 2022; 12:1014897. [DOI: 10.3389/fcimb.2022.1014897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Rv0790c is predicted to be a conserved hypothetical protein encoded by Mycobacterium tuberculosis (Mtb). However, its function in Mtb infection remains largely unknown. In this study, we found that Rv0790c promoted bacillary survival of M. smegmatis (Ms), both in vitro and in vivo. The bacillary burden of Ms exogenously expressing Rv0790c increased, whereas in Rv0790c-knockouts the bacillary burden decreased in infected macrophages. Multiple cellular processes were analyzed to explore the underlying mechanisms. We found that neither inflammatory regulation nor apoptotic induction were responsible for the promotion of bacillary survival mediated by Rv0790c. Interestingly, we found that Rv0790c facilitates mycobacterial survival through cellular autophagy at its early stage. Immunoprecipitation assay of autophagy initiation-related proteins indicated that Rv0790c interacted with mTOR and enhanced its activity, as evidenced by the increased phosphorylation level of mTOR downstream substrates, ULK-1, at Ser757 and P70S6K, at Thr389. Our study uncovers a novel autophagy suppressor encoded by mycobacterial Rv0790c, which inhibits the early stage of cellular autophagy induction upon Mtb infection and takes an important role in maintaining intracellular mycobacterial survival. It may aid in understanding the mechanism of Mtb evasion of host cellular degradation, as well as hold the potential to develop new targets for the prevention and treatment of tuberculosis.
Collapse
|
12
|
Gough M, Singh DK, Singh B, Kaushal D, Mehra S. System-wide identification of myeloid markers of TB disease and HIV-induced reactivation in the macaque model of Mtb infection and Mtb/SIV co-infection. Front Immunol 2022; 13:777733. [PMID: 36275677 PMCID: PMC9583676 DOI: 10.3389/fimmu.2022.777733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) has developed specialized mechanisms to parasitize its host cell, the macrophage. These mechanisms allow it to overcome killing by oxidative burst and persist in the wake of an inflammatory response. Mtb infection in the majority of those exposed is controlled in an asymptomatic form referred to as latent tuberculosis infection (LTBI). HIV is a well-known catalyst of reactivation of LTBI to active TB infection (ATB). Through the use of nonhuman primates (NHPs) co-infected with Mtb and Simian Immunodeficiency Virus (Mtb/SIV), we are able to simulate human progression of TB/AIDS comorbidity. The advantage of NHP models is that they recapitulate the breadth of human TB outcomes, including immune control of infection, and loss of this control due to SIV co-infection. Identifying correlates of immune control of infection is important for both vaccine and therapeutics development. Using macaques infected with Mtb or Mtb/SIV and with different clinical outcomes we attempted to identify signatures between those that progress to active infection after SIV challenge (reactivators) and those that control the infection (non-reactivators). We particularly focused on pathways relevant to myeloid origin cells such as macrophages, as these innate immunocytes have an important contribution to the initial control or the lack thereof, following Mtb infection. Using bacterial burden, C-reactive protein (CRP), and other clinical indicators of disease severity as a guide, we were able to establish gene signatures of host disease state and progression. In addition to gene signatures, clustering algorithms were used to differentiate between host disease states and identify relationships between genes. This allowed us to identify clusters of genes which exhibited differential expression profiles between the three groups of macaques: ATB, LTBI and Mtb/SIV. The gene signatures were associated with pathways relevant to apoptosis, ATP production, phagocytosis, cell migration, and Type I interferon (IFN), which are related to macrophage function. Our results suggest novel macrophage functions that may play roles in the control of Mtb infection with and without co-infection with SIV. These results particularly point towards an interplay between Type I IFN signaling and IFN-γ signaling, and the resulting impact on lung macrophages as an important determinant of progression to TB.
Collapse
Affiliation(s)
| | | | | | | | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
13
|
Martínez-Pérez A, Estévez O, González-Fernández Á. Contribution and Future of High-Throughput Transcriptomics in Battling Tuberculosis. Front Microbiol 2022; 13:835620. [PMID: 35283833 PMCID: PMC8908424 DOI: 10.3389/fmicb.2022.835620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/25/2022] [Indexed: 11/13/2022] Open
Abstract
While Tuberculosis (TB) infection remains a serious challenge worldwide, big data and “omic” approaches have greatly contributed to the understanding of the disease. Transcriptomics have been used to tackle a wide variety of queries including diagnosis, treatment evolution, latency and reactivation, novel target discovery, vaccine response or biomarkers of protection. Although a powerful tool, the elevated cost and difficulties in data interpretation may hinder transcriptomics complete potential. Technology evolution and collaborative efforts among multidisciplinary groups might be key in its exploitation. Here, we discuss the main fields explored in TB using transcriptomics, and identify the challenges that need to be addressed for a real implementation in TB diagnosis, prevention and therapy.
Collapse
Affiliation(s)
- Amparo Martínez-Pérez
- Biomedical Research Center (CINBIO), Universidade de Vigo, Vigo, Spain.,Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-GS), Vigo, Spain
| | - Olivia Estévez
- Biomedical Research Center (CINBIO), Universidade de Vigo, Vigo, Spain.,Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-GS), Vigo, Spain
| | - África González-Fernández
- Biomedical Research Center (CINBIO), Universidade de Vigo, Vigo, Spain.,Hospital Álvaro Cunqueiro, Galicia Sur Health Research Institute (IIS-GS), Vigo, Spain
| |
Collapse
|
14
|
Willemse D, Moodley C, Mehra S, Kaushal D. Transcriptional Response of Mycobacterium tuberculosis to Cigarette Smoke Condensate. Front Microbiol 2021; 12:744800. [PMID: 34721344 PMCID: PMC8554204 DOI: 10.3389/fmicb.2021.744800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
Smoking is known to be an added risk factor for tuberculosis (TB), with nearly a quarter of the TB cases attributed to cigarette smokers in the 22 countries with the highest TB burden. Many studies have indicated a link between risk of active TB and cigarette smoke. Smoking is also known to significantly decrease TB cure and treatment completion rate and increase mortality rates. Cigarette smoke contains thousands of volatile compounds including carcinogens, toxins, reactive solids, and oxidants in both particulate and gaseous phase. Yet, to date, limited studies have analyzed the impact of cigarette smoke components on Mycobacterium tuberculosis (Mtb), the causative agent of TB. Here we report the impact of cigarette smoke condensate (CSC) on survival, mutation frequency, and gene expression of Mtb in vitro. We show that exposure of virulent Mtb to cigarette smoke increases the mutation frequency of the pathogen and strongly induces the expression of the regulon controlled by SigH—a global transcriptional regulator of oxidative stress. SigH has previously been shown to be required for Mtb to respond to oxidative stress, survival, and granuloma formation in vivo. A high-SigH expression phenotype is known to be associated with greater virulence of Mtb. In patients with pulmonary TB who smoke, these changes may therefore play an important, yet unexplored, role in the treatment efficacy by potentially enhancing the virulence of tubercle bacilli.
Collapse
Affiliation(s)
- Danicke Willemse
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Chivonne Moodley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States.,Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States
| | - Smriti Mehra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States.,Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
15
|
Sharma S, Sharma M. Proline-Glutamate/Proline-Proline-Glutamate (PE/PPE) proteins of Mycobacterium tuberculosis: The multifaceted immune-modulators. Acta Trop 2021; 222:106035. [PMID: 34224720 DOI: 10.1016/j.actatropica.2021.106035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/30/2022]
Abstract
The PE/PPE proteins encoded by seven percent (7%) of Mycobacterium tuberculosis (Mtb) genome are the chief constituents to pathogen's virulence reservoir. The fact that these genes have evolved along ESX secretory system in pathogenic Mtb strains make their investigation very intriguing. There is lot of speculation about the prominent role of these proteins at host pathogen interface and in disease pathogenesis. Nevertheless, the exact function of PE/PPE proteins still remains a mystery which calls for further research targeting these proteins. This article is an effort to document all the facts known so far with regard to these unique proteins which involves their origin, evolution, transcriptional control, and most important their role as host immune-modulators. Our understanding strongly points towards the versatile nature of these PE/PPE proteins as Mtb's host immune sensors and as decisive factors in shaping the outcome of infection. Further investigation on these proteins will surely pave way for newer and effective vaccines and therapeutics to control Tuberculosis (TB).
Collapse
Affiliation(s)
- Sadhna Sharma
- DS Kothari Central Interdisciplinary Research Centre and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| | - Monika Sharma
- DS Kothari Central Interdisciplinary Research Centre and Department of Zoology, Miranda House, University of Delhi, Delhi 110007, India.
| |
Collapse
|
16
|
Forrellad MA, Vázquez CL, Blanco FC, Klepp LI, García EA, Rocha RV, Luciana V, Bigi MM, Gutierrez MG, Bigi F. Rv2617c and P36 are virulence factors of pathogenic mycobacteria involved in resistance to oxidative stress. Virulence 2019; 10:1026-1033. [PMID: 31782338 PMCID: PMC6930017 DOI: 10.1080/21505594.2019.1693714] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study, we characterized the role of Rv2617c in the virulence of Mycobacterium tuberculosis. Rv2617c is a protein of unknown function unique to M. tuberculosis complex (MTC) and Mycobacterium leprae. In vitro, this protein interacts with the virulence factor P36 (also named Erp) and KdpF, a protein linked to nitrosative stress. Here, we showed that knockout of the Rv2617c gene in M. tuberculosis CDC1551 reduced the replication of the pathogen in a mouse model of infection and favored the trafficking of mycobacteria to phagolysosomes. We also demonstrated that Rv2617c and P36 are required for resistance to in vitro hydrogen peroxide treatment in M. tuberculosis and Mycobacterium bovis, respectively. These findings indicate Rv2617c and P36 act in concert to prevent bacterial damage upon oxidative stress.
Collapse
Affiliation(s)
- Marina A Forrellad
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Cristina L Vázquez
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Federico C Blanco
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Laura I Klepp
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Elizabeth A García
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Rosana V Rocha
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - Villafañe Luciana
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| | - María M Bigi
- University of Buenos Aires, School of Agronomy (Universidad de Buenos Aires, Facultad de Agronomía), Autonomous City of Bueno Aires, Argentine
| | - Maximiliano G Gutierrez
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, London, UK
| | - Fabiana Bigi
- Institute of Biotechnology, National Institute of Agricultural Technology (INTA, Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) and IABIMO-National Scientific and Technical Research Council (CONICET, Consejo Nacional de Investigaciones Científicas y Tecnológicas), Hurlingham, Buenos Aires, Argentine
| |
Collapse
|
17
|
Anthony RM, den Hertog AL, van Soolingen D. 'Happy the man, who, studying nature's laws, Thro' known effects can trace the secret cause.' Do we have enough pieces to solve the pyrazinamide puzzle? J Antimicrob Chemother 2019. [PMID: 29528413 DOI: 10.1093/jac/dky060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A low pH was assumed to be required for the activity of pyrazinoic acid (the active form of pyrazinamide) against Mycobacterium tuberculosis, but recently activity has been demonstrated at neutral pH. Renewed interest in pyrazinamide has led to an increasing number of potential targets and the suspicion that pyrazinamide is a 'dirty drug'. However, it is our opinion that the recent demonstration that pyrazinoic acid is active against PanD provides an alternative explanation for the secret of pyrazinamide's unusual activity. In this article we propose that PanD is the primary target of pyrazinoic acid but expression of pyrazinoic acid susceptibility requires an intact stress response. As the mycobacterial stress response requires the interaction of a number of genes, disruption of any could result in an inability to enter the susceptible phenotype. We believe this model can explain most of the recent observations of the seemingly diverse spectrum of activity of pyrazinamide.
Collapse
Affiliation(s)
- R M Anthony
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - A L den Hertog
- Institute for Life Sciences and Chemistry, HU University of Applied Sciences Utrecht, Utrecht, The Netherlands
| | - D van Soolingen
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
18
|
Safi H, Gopal P, Lingaraju S, Ma S, Levine C, Dartois V, Yee M, Li L, Blanc L, Ho Liang HP, Husain S, Hoque M, Soteropoulos P, Rustad T, Sherman DR, Dick T, Alland D. Phase variation in Mycobacterium tuberculosis glpK produces transiently heritable drug tolerance. Proc Natl Acad Sci U S A 2019; 116:19665-19674. [PMID: 31488707 PMCID: PMC6765255 DOI: 10.1073/pnas.1907631116] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The length and complexity of tuberculosis (TB) therapy, as well as the propensity of Mycobacterium tuberculosis to develop drug resistance, are major barriers to global TB control efforts. M. tuberculosis is known to have the ability to enter into a drug-tolerant state, which may explain many of these impediments to TB treatment. We have identified a mechanism of genetically encoded but rapidly reversible drug tolerance in M. tuberculosis caused by transient frameshift mutations in a homopolymeric tract (HT) of 7 cytosines (7C) in the glpK gene. Inactivating frameshift mutations associated with the 7C HT in glpK produce small colonies that exhibit heritable multidrug increases in minimal inhibitory concentrations and decreases in drug-dependent killing; however, reversion back to a fully drug-susceptible large-colony phenotype occurs rapidly through the introduction of additional insertions or deletions in the same glpK HT region. These reversible frameshift mutations in the 7C HT of M. tuberculosis glpK occur in clinical isolates, accumulate in M. tuberculosis-infected mice with further accumulation during drug treatment, and exhibit a reversible transcriptional profile including induction of dosR and sigH and repression of kstR regulons, similar to that observed in other in vitro models of M. tuberculosis tolerance. These results suggest that GlpK phase variation may contribute to drug tolerance, treatment failure, and relapse in human TB. Drugs effective against phase-variant M. tuberculosis may hasten TB treatment and improve cure rates.
Collapse
Affiliation(s)
- Hassan Safi
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103;
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Pooja Gopal
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Republic of Singapore
| | - Subramanya Lingaraju
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Shuyi Ma
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - Carly Levine
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Veronique Dartois
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Michelle Yee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Republic of Singapore
| | - Liping Li
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Landry Blanc
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Hsin-Pin Ho Liang
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Seema Husain
- Genomics Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Mainul Hoque
- Genomics Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | | | - Tige Rustad
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - David R Sherman
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - David Alland
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103;
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| |
Collapse
|
19
|
Gautam US, Mehra S, Kumari P, Alvarez X, Niu T, Tyagi JS, Kaushal D. Mycobacterium tuberculosis sensor kinase DosS modulates the autophagosome in a DosR-independent manner. Commun Biol 2019; 2:349. [PMID: 31552302 PMCID: PMC6754383 DOI: 10.1038/s42003-019-0594-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 09/03/2019] [Indexed: 01/03/2023] Open
Abstract
Dormancy is a key characteristic of the intracellular life-cycle of Mtb. The importance of sensor kinase DosS in mycobacteria are attributed in part to our current findings that DosS is required for both persistence and full virulence of Mtb. Here we show that DosS is also required for optimal replication in macrophages and involved in the suppression of TNF-α and autophagy pathways. Silencing of these pathways during the infection process restored full virulence in MtbΔdosS mutant. Notably, a mutant of the response regulator DosR did not exhibit the attenuation in macrophages, suggesting that DosS can function independently of DosR. We identified four DosS targets in Mtb genome; Rv0440, Rv2859c, Rv0994, and Rv0260c. These genes encode functions related to hypoxia adaptation, which are not directly controlled by DosR, e.g., protein recycling and chaperoning, biosynthesis of molybdenum cofactor and nitrogen metabolism. Our results strongly suggest a DosR-independent role for DosS in Mtb.
Collapse
Affiliation(s)
- Uma S. Gautam
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Present Address: Duke Human Vaccine Institute, Duke University School of Medicine, 909 S. LaSalle St., Durham, NC 27710 USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803 USA
- Center for Experimental Infectious Diseases Research, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803 USA
| | - Priyanka Kumari
- All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Xavier Alvarez
- Tulane National Primate Research Center, Covington, LA 70433 USA
| | - Tianhua Niu
- Department of Biochemistry, Tulane University School of Medicine, New Orleans, 70112 LA USA
| | - Jaya S. Tyagi
- All India Institute of Medical Sciences, New Delhi, 110029 India
- Centre for Bio-design and Diagnostics, Translational Health Science and Technology Institute Faridabad, Haryana, 121001 India
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433 USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, 70112 LA USA
| |
Collapse
|
20
|
Reyes AM, Pedre B, De Armas MI, Tossounian MA, Radi R, Messens J, Trujillo M. Chemistry and Redox Biology of Mycothiol. Antioxid Redox Signal 2018; 28:487-504. [PMID: 28372502 DOI: 10.1089/ars.2017.7074] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
SIGNIFICANCE Mycothiol (MSH, AcCys-GlcN-Ins) is the main low-molecular weight (LMW) thiol of most Actinomycetes, including the human pathogen Mycobacterium tuberculosis that affects millions of people worldwide. Strains with decreased MSH content show increased susceptibilities to hydroperoxides and electrophilic compounds. In M. tuberculosis, MSH modulates the response to several antituberculosis drugs. Enzymatic routes involving MSH could provide clues for specific drug design. Recent Advances: Physicochemical data argue against a rapid, nonenzymatic reaction of MSH with oxidants, disulfides, or electrophiles. Moreover, exposure of the bacteria to high concentrations of two-electron oxidants resulted in protein mycothiolation. The recently described glutaredoxin-like protein mycoredoxin-1 (Mrx-1) provides a route for catalytic reduction of mycothiolated proteins, protecting critical cysteines from irreversible oxidation. The description of MSH/Mrx-1-dependent activities of peroxidases helped to explain the higher susceptibility to oxidants observed in Actinomycetes lacking MSH. Moreover, the first mycothiol-S-transferase, member of the DinB superfamily of proteins, was described. In Corynebacterium, both the MSH/Mrx-1 and the thioredoxin pathways reduce methionine sulfoxide reductase A. A novel tool for in vivo imaging of the MSH/mycothiol disulfide (MSSM) status allows following changes in the mycothiol redox state during macrophage infection and its relationship with antibiotic sensitivity. CRITICAL ISSUES Redundancy of MSH with other LMW thiols is starting to be unraveled and could help to rationalize the differences in the reported importance of MSH synthesis observed in vitro versus in animal infection models. FUTURE DIRECTIONS Future work should be directed to establish the structural bases of the specificity of MSH-dependent enzymes, thus facilitating drug developments. Antioxid. Redox Signal. 28, 487-504.
Collapse
Affiliation(s)
- Aníbal M Reyes
- 1 Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Montevideo, Uruguay .,2 Center for Free Radical and Biomedical Research , Universidad de la República, Montevideo, Uruguay
| | - Brandán Pedre
- 3 Center for Structural Biology , VIB, Brussels, Belgium .,4 Brussels Center for Redox Biology , Brussels, Belgium .,5 Structural Biology Brussels, Vrije Universiteit Brussel , Brussels, Belgium
| | - María Inés De Armas
- 1 Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Montevideo, Uruguay .,2 Center for Free Radical and Biomedical Research , Universidad de la República, Montevideo, Uruguay
| | - Maria-Armineh Tossounian
- 3 Center for Structural Biology , VIB, Brussels, Belgium .,4 Brussels Center for Redox Biology , Brussels, Belgium .,5 Structural Biology Brussels, Vrije Universiteit Brussel , Brussels, Belgium
| | - Rafael Radi
- 1 Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Montevideo, Uruguay .,2 Center for Free Radical and Biomedical Research , Universidad de la República, Montevideo, Uruguay
| | - Joris Messens
- 3 Center for Structural Biology , VIB, Brussels, Belgium .,4 Brussels Center for Redox Biology , Brussels, Belgium .,5 Structural Biology Brussels, Vrije Universiteit Brussel , Brussels, Belgium
| | - Madia Trujillo
- 1 Departamento de Bioquímica, Facultad de Medicina, Universidad de la República , Montevideo, Uruguay .,2 Center for Free Radical and Biomedical Research , Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
21
|
Foreman TW, Veatch AV, LoBato DN, Didier PJ, Doyle-Meyers LA, Russell-Lodrigue KE, Lackner AA, Kousoulas KG, Khader SA, Kaushal D, Mehra S. Nonpathologic Infection of Macaques by an Attenuated Mycobacterial Vaccine Is Not Reactivated in the Setting of HIV Co-Infection. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2811-2820. [PMID: 28935575 PMCID: PMC5718104 DOI: 10.1016/j.ajpath.2017.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/29/2022]
Abstract
Failure to replace Bacille Calmette-Guerin vaccines with efficacious anti-tuberculosis (TB) vaccines have prompted outside-the-box thinking, including pulmonary vaccination to elicit local immunity. Inhalational MtbΔsigH, a stress-response-attenuated strain, protected against lethal TB in macaques. While live mycobacterial vaccines show promising efficacy, HIV co-infection and the resulting immunodeficiency prompts safety concerns about their use. We assessed the persistence and safety of MtbΔsigH, delivered directly to the lungs, in the setting of HIV co-infection. Macaques were aerosol-vaccinated with ΔsigH and subsequently challenged with SIVmac239. Bronchoalveolar lavage and tissues were sampled for mycobacterial persistence, pathology, and immune correlates. Only 35% and 3.5% of lung samples were positive for live bacilli and granulomas, respectively. Our results therefore suggest that the nonpathologic infection of macaque lungs by ΔsigH was not reactivated by simian immunodeficiency virus, despite high viral levels and massive ablation of pulmonary CD4+ T cells. Protective pulmonary responses were retained, including vaccine-induced bronchus-associated lymphoid tissue and CD8+ effector memory T cells. Despite acute simian immunodeficiency virus infection, all animals remained asymptomatic of pulmonary TB. These findings highlight the efficacy of mucosal vaccination via this attenuated strain and will guide its further development to potentially combat TB in HIV-endemic areas. Our results also suggest that a lack of pulmonary pathology is a key correlate of the safety of live mycobacterial vaccines.
Collapse
Affiliation(s)
- Taylor W Foreman
- Tulane National Primate Research Center, Covington, Louisiana; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ashley V Veatch
- Tulane National Primate Research Center, Covington, Louisiana
| | - Denae N LoBato
- Tulane National Primate Research Center, Covington, Louisiana
| | - Peter J Didier
- Tulane National Primate Research Center, Covington, Louisiana
| | | | | | - Andrew A Lackner
- Tulane National Primate Research Center, Covington, Louisiana; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Konstantin G Kousoulas
- Center for Biomedical Research Excellence, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana; Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, Louisiana; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana.
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, Louisiana; Center for Biomedical Research Excellence, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana; Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana.
| |
Collapse
|
22
|
Veatch AV, Kaushal D. Opening Pandora's Box: Mechanisms of Mycobacterium tuberculosis Resuscitation. Trends Microbiol 2017; 26:145-157. [PMID: 28911979 DOI: 10.1016/j.tim.2017.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/12/2017] [Accepted: 08/03/2017] [Indexed: 12/27/2022]
Abstract
Mycobacterium tuberculosis (Mtb) characteristically causes an asymptomatic infection. While this latent tuberculosis infection (LTBI) is not contagious, reactivation to active tuberculosis disease (TB) causes the patient to become infectious. A vaccine has existed for TB for a century, while drug treatments have been available for over 70 years; despite this, TB remains a major global health crisis. Understanding the factors which allow the bacillus to control responses to host stress and mechanisms leading to latency are critical for persistence. Similarly, molecular switches which respond to reactivation are important. Recently, research in the field has sought to focus on reactivation, employing system-wide approaches and animal models. Here, we describe the current work that has been done to elucidate the mechanisms of reactivation and stop reactivation in its tracks.
Collapse
Affiliation(s)
- Ashley V Veatch
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Deepak Kaushal
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
23
|
Hudock TA, Foreman TW, Bandyopadhyay N, Gautam US, Veatch AV, LoBato DN, Gentry KM, Golden NA, Cavigli A, Mueller M, Hwang SA, Hunter RL, Alvarez X, Lackner AA, Bader JS, Mehra S, Kaushal D. Hypoxia Sensing and Persistence Genes Are Expressed during the Intragranulomatous Survival of Mycobacterium tuberculosis. Am J Respir Cell Mol Biol 2017; 56:637-647. [PMID: 28135421 PMCID: PMC5449490 DOI: 10.1165/rcmb.2016-0239oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022] Open
Abstract
Although it is accepted that the environment within the granuloma profoundly affects Mycobacterium tuberculosis (Mtb) and infection outcome, our ability to understand Mtb gene expression in these niches has been limited. We determined intragranulomatous gene expression in human-like lung lesions derived from nonhuman primates with both active tuberculosis (ATB) and latent TB infection (LTBI). We employed a non-laser-based approach to microdissect individual lung lesions and interrogate the global transcriptome of Mtb within granulomas. Mtb genes expressed in classical granulomas with central, caseous necrosis, as well as within the caseum itself, were identified and compared with other Mtb lesions in animals with ATB (n = 7) or LTBI (n = 7). Results were validated using both an oligonucleotide approach and RT-PCR on macaque samples and by using human TB samples. We detected approximately 2,900 and 1,850 statistically significant genes in ATB and LTBI lesions, respectively (linear models for microarray analysis, Bonferroni corrected, P < 0.05). Of these genes, the expression of approximately 1,300 (ATB) and 900 (LTBI) was positively induced. We identified the induction of key regulons and compared our results to genes previously determined to be required for Mtb growth. Our results indicate pathways that Mtb uses to ensure its survival in a highly stressful environment in vivo. A large number of genes is commonly expressed in granulomas with ATB and LTBI. In addition, the enhanced expression of the dormancy survival regulon was a key feature of lesions in animals with LTBI, stressing its importance in the persistence of Mtb during the chronic phase of infection.
Collapse
Affiliation(s)
- Teresa A. Hudock
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane University Health Sciences, New Orleans, Louisiana; and
| | - Taylor W. Foreman
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane University Health Sciences, New Orleans, Louisiana; and
| | - Nirmalya Bandyopadhyay
- Whitaker Biomedical Engineering Institute, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Uma S. Gautam
- Tulane National Primate Research Center, Covington, Louisiana
| | - Ashley V. Veatch
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane University Health Sciences, New Orleans, Louisiana; and
| | - Denae N. LoBato
- Tulane National Primate Research Center, Covington, Louisiana
| | | | - Nadia A. Golden
- Tulane National Primate Research Center, Covington, Louisiana
| | - Amy Cavigli
- Tulane National Primate Research Center, Covington, Louisiana
| | | | - Shen-An Hwang
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, Houston, Texas
| | - Robert L. Hunter
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, Houston, Texas
| | - Xavier Alvarez
- Tulane National Primate Research Center, Covington, Louisiana
| | - Andrew A. Lackner
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane University Health Sciences, New Orleans, Louisiana; and
| | - Joel S. Bader
- Whitaker Biomedical Engineering Institute, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Smriti Mehra
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, Louisiana
- Tulane University Health Sciences, New Orleans, Louisiana; and
| |
Collapse
|
24
|
Abshire CF, Prasai K, Soto I, Shi R, Concha M, Baddoo M, Flemington EK, Ennis DG, Scott RS, Harrison L. Exposure of Mycobacterium marinum to low-shear modeled microgravity: effect on growth, the transcriptome and survival under stress. NPJ Microgravity 2016; 2:16038. [PMID: 28725743 PMCID: PMC5515531 DOI: 10.1038/npjmgrav.2016.38] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 09/15/2016] [Accepted: 09/17/2016] [Indexed: 12/04/2022] Open
Abstract
Waterborne pathogenic mycobacteria can form biofilms, and certain species can cause hard-to-treat human lung infections. Astronaut health could therefore be compromised if the spacecraft environment or water becomes contaminated with pathogenic mycobacteria. This work uses Mycobacterium marinum to determine the physiological changes in a pathogenic mycobacteria grown under low-shear modeled microgravity (LSMMG). M. marinum were grown in high aspect ratio vessels (HARVs) using a rotary cell culture system subjected to LSMMG or the control orientation (normal gravity, NG) and the cultures used to determine bacterial growth, bacterium size, transcriptome changes, and resistance to stress. Two exposure times to LSMMG and NG were examined: bacteria were grown for ~40 h (short), or 4 days followed by re-dilution and growth for ~35 h (long). M. marinum exposed to LSMMG transitioned from exponential phase earlier than the NG culture. They were more sensitive to hydrogen peroxide but showed no change in resistance to gamma radiation or pH 3.5. RNA-Seq detected significantly altered transcript levels for 562 and 328 genes under LSMMG after short and long exposure times, respectively. Results suggest that LSMMG induced a reduction in translation, a downregulation of metabolism, an increase in lipid degradation, and increased chaperone and mycobactin expression. Sigma factor H (sigH) was the only sigma factor transcript induced by LSMMG after both short and long exposure times. In summary, transcriptome studies suggest that LSMMG may simulate a nutrient-deprived environment similar to that found within macrophage during infection. SigH is also implicated in the M. marinum LSMMG transcriptome response.
Collapse
Affiliation(s)
- Camille F Abshire
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Kanchanjunga Prasai
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Israel Soto
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Runhua Shi
- Department of Medicine and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Monica Concha
- Department of Pathology and Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology and Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology and Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Don G Ennis
- Department of Biology, University of Louisiana, Lafayette, LA, USA
| | - Rona S Scott
- Department of Microbiology and Immunology, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Lynn Harrison
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
25
|
Venkatesan A, Palaniyandi K, Sharma D, Bisht D, Narayanan S. Functional Characterization of PknI-Rv2159c Interaction in Redox Homeostasis of Mycobacterium tuberculosis. Front Microbiol 2016; 7:1654. [PMID: 27818650 PMCID: PMC5073100 DOI: 10.3389/fmicb.2016.01654] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/04/2016] [Indexed: 01/02/2023] Open
Abstract
Mycobacterium tuberculosis adapts to stress conditions by responding to the signals from its external environment. M. tuberculosis genome encodes 11 eukaryotic like serine/threonine protein kinases (STPK) and their importance in regulating the physiology and virulence of the bacteria are being explored. Previous study from our lab identified the M. tuberculosis STPK, PknI interacts with two peroxidase proteins such as Rv2159c and Rv0148. In this study, we have characterized the biological function behind the PknI-Rv2159c interaction in M. tuberculosis. Point mutation of Ala-Gly-Trp motif identified that only Ala49 and Gly50 amino acids of Rv2159c are responsible for interaction and there is no phosphorylation involved in the PknI-Rv2159c interaction. Rv2159c is a member from the carboxymuconolactone decarboxylase family with peroxidase activity. Enzymatic assays with catalytic site point mutants showed that Cys84 of Rv2159c was responsible for its alkylhydroperoxidase activity. Interestingly, interaction with PknI increased its peroxidase activity by several folds. Gene knockdown of Rv2159c in M. tuberculosis showed increased sensitivity to peroxides such as cumene hydroperoxide and hydrogen peroxide. Proteomic analysis of differentially expressing Rv2159c strains by 2D gel electrophoresis and mass spectrometry revealed the differential abundance of 21 proteins. The total absence of oxidoreductase, GuaB1 suggests the essential role of Rv2159c in redox maintenance. Our findings provide new insights on signaling mechanisms of PknI in maintaining the redox homeostasis during oxidative stresses.
Collapse
Affiliation(s)
- Arunkumar Venkatesan
- Department of Immunology, National Institute for Research in TuberculosisChennai, India
| | - Kannan Palaniyandi
- Department of Immunology, National Institute for Research in TuberculosisChennai, India
| | - Divakar Sharma
- Department of Biochemistry, National JALMA Institute for Leprosy and other Mycobacterial DiseasesAgra, India
| | - Deepa Bisht
- Department of Biochemistry, National JALMA Institute for Leprosy and other Mycobacterial DiseasesAgra, India
| | - Sujatha Narayanan
- Department of Immunology, National Institute for Research in TuberculosisChennai, India
| |
Collapse
|
26
|
Veatch AV, Niu T, Caskey J, McGillivray A, Gautam US, Subramanian R, Kousoulas KG, Mehra S, Kaushal D. Sequencing-relative to hybridization-based transcriptomics approaches better define Mycobacterium tuberculosis stress-response regulons. Tuberculosis (Edinb) 2016; 101S:S9-S17. [PMID: 27729257 DOI: 10.1016/j.tube.2016.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mycobacterium tuberculosis (Mtb) infections cause tuberculosis (TB), an infectious disease which causes ∼1.5 million deaths annually. The ability of this pathogen to evade, escape and encounter immune surveillance is fueled by its adaptability. Thus, Mtb induces a transition in its transcriptome in response to environmental changes. Global transcriptome profiling has been key to our understanding of how Mtb responds to the different stress conditions it faces during its life cycle. While this was initially achieved using microarray technology, RNAseq is now widely employed. It is important to understand the correlation between the large amount of microarray based transcriptome data, which continues to shape our understanding of Mtb stress networks, and newer data being generated using RNAseq. We assessed how well the two platforms correlate using three well-defined stress conditions: diamide, hypoxia, and re-aeration. The data used here was generated by different individuals over time using distinct samples, providing a stringent test of platform correlation. While correlation between microarrays and sequencing was high upon diamide treatment, which causes a rapid reprogramming of the transcriptome, RNAseq allowed a better definition of the hypoxic response, characterized by subtle changes in the magnitude of gene-expression. RNAseq also allows for the best cross-platform reproducibility.
Collapse
Affiliation(s)
- Ashley V Veatch
- Divisions of Bacteriology & Parasitology, Tulane National Primate Research Center, Covington LA, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tianhua Niu
- Department of Biostatistics & Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans LA, USA
| | - John Caskey
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Amanda McGillivray
- Divisions of Bacteriology & Parasitology, Tulane National Primate Research Center, Covington LA, USA
| | - Uma Shankar Gautam
- Divisions of Bacteriology & Parasitology, Tulane National Primate Research Center, Covington LA, USA
| | - Ramesh Subramanian
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - K Gus Kousoulas
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Smriti Mehra
- Divisions of Microbiology, Tulane National Primate Research Center, Covington LA, USA; Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, USA
| | - Deepak Kaushal
- Divisions of Bacteriology & Parasitology, Tulane National Primate Research Center, Covington LA, USA; Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
27
|
Sigma Factors: Key Molecules in Mycobacterium tuberculosis Physiology and Virulence. Microbiol Spectr 2015; 2:MGM2-0007-2013. [PMID: 26082107 DOI: 10.1128/microbiolspec.mgm2-0007-2013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rapid adaptation to changing environments is one of the keys to the success of microorganisms. Since infection is a dynamic process, it is possible to predict that Mycobacterium tuberculosis adaptation involves continuous modulation of its global transcriptional profile in response to the changing environment found in the human body. In the last 18 years several studies have stressed the role of sigma (σ) factors in this process. These are small interchangeable subunits of the RNA polymerase holoenzyme that are required for transcriptional initiation and that determine promoter specificity. The M. tuberculosis genome encodes 13 of these proteins, one of which--the principal σ factor σA--is essential. Of the other 12 σ factors, at least 6 are required for virulence. In this article we review our current knowledge of mycobacterial σ factors, their regulons, the complex mechanisms determining their regulation, and their roles in M. tuberculosis physiology and virulence.
Collapse
|
28
|
Kaushal D, Foreman TW, Gautam US, Alvarez X, Adekambi T, Rangel-Moreno J, Golden NA, Johnson AMF, Phillips BL, Ahsan MH, Russell-Lodrigue KE, Doyle LA, Roy CJ, Didier PJ, Blanchard JL, Rengarajan J, Lackner AA, Khader SA, Mehra S. Mucosal vaccination with attenuated Mycobacterium tuberculosis induces strong central memory responses and protects against tuberculosis. Nat Commun 2015; 6:8533. [PMID: 26460802 PMCID: PMC4608260 DOI: 10.1038/ncomms9533] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/02/2015] [Indexed: 02/07/2023] Open
Abstract
Tuberculosis (TB) is a global pandaemic, partially due to the failure of vaccination approaches. Novel anti-TB vaccines are therefore urgently required. Here we show that aerosol immunization of macaques with the Mtb mutant in SigH (MtbΔsigH) results in significant recruitment of inducible bronchus-associated lymphoid tissue (iBALT) as well as CD4+ and CD8+ T cells expressing activation and proliferation markers to the lungs. Further, the findings indicate that pulmonary vaccination with MtbΔsigH elicited strong central memory CD4+ and CD8+ T-cell responses in the lung. Vaccination with MtbΔsigH results in significant protection against a lethal TB challenge, as evidenced by an approximately three log reduction in bacterial burdens, significantly diminished clinical manifestations and granulomatous pathology and characterized by the presence of profound iBALT. This highly protective response is virtually absent in unvaccinated and BCG-vaccinated animals after challenge. These results suggest that future TB vaccine candidates can be developed on the basis of MtbΔsigH. BCG, the only vaccine currently used against tuberculosis, confers only limited protection. Here the authors show that mucosal immunization of macaques with an attenuated strain of Mycobacterium tuberculosis confers a high level of protection from a lethal challenge with the bacterium.
Collapse
Affiliation(s)
- Deepak Kaushal
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.,Department of Microbiology and Immunology, Tulane Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Taylor W Foreman
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.,Biomedical Sciences Graduate Program, Tulane Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Uma S Gautam
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - Xavier Alvarez
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - Toidi Adekambi
- Yerkes National Primate Research Center, Atlanta, Georgia 30329, USA.,Emory Vaccine Center, Atlanta, Georgia 30329, USA
| | | | - Nadia A Golden
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | | | - Bonnie L Phillips
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.,Biomedical Sciences Graduate Program, Tulane Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Muhammad H Ahsan
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | | | - Lara A Doyle
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - Chad J Roy
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - Peter J Didier
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - James L Blanchard
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA
| | - Jyothi Rengarajan
- Yerkes National Primate Research Center, Atlanta, Georgia 30329, USA.,Emory Vaccine Center, Atlanta, Georgia 30329, USA
| | - Andrew A Lackner
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.,Department of Microbiology and Immunology, Tulane Health Sciences Center, New Orleans, Louisiana 70112, USA.,Department of Pathology, Tulane Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University at St Louis, St Louis, Missouri 63110, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.,Center for Biomedical Research Excellence, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana 70803, USA.,Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana 70803, USA
| |
Collapse
|
29
|
Mothé BR, Lindestam Arlehamn CS, Dow C, Dillon MBC, Wiseman RW, Bohn P, Karl J, Golden NA, Gilpin T, Foreman TW, Rodgers MA, Mehra S, Scriba TJ, Flynn JL, Kaushal D, O'Connor DH, Sette A. The TB-specific CD4(+) T cell immune repertoire in both cynomolgus and rhesus macaques largely overlap with humans. Tuberculosis (Edinb) 2015; 95:722-735. [PMID: 26526557 DOI: 10.1016/j.tube.2015.07.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/23/2015] [Accepted: 07/26/2015] [Indexed: 01/05/2023]
Abstract
Non-human primate (NHP) models of tuberculosis (TB) immunity and pathogenesis, especially rhesus and cynomolgus macaques, are particularly attractive because of the high similarity of the human and macaque immune systems. However, little is known about the MHC class II epitopes recognized in macaques, thus hindering the establishment of immune correlates of immunopathology and protective vaccination. We characterized immune responses in rhesus macaques vaccinated against and/or infected with Mycobacterium tuberculosis (Mtb), to a panel of antigens currently in human vaccine trials. We defined 54 new immunodominant CD4(+) T cell epitopes, and noted that antigens immunodominant in humans are also immunodominant in rhesus macaques, including Rv3875 (ESAT-6) and Rv3874 (CFP10). Pedigree and inferred restriction analysis demonstrated that this phenomenon was not due to common ancestry or inbreeding, but rather presentation by common alleles, as well as, promiscuous binding. Experiments using a second cohort of rhesus macaques demonstrated that a pool of epitopes defined in the previous experiments can be used to detect T cell responses in over 75% of individual monkeys. Additionally, 100% of cynomolgus macaques, irrespective of their latent or active TB status, responded to rhesus and human defined epitope pools. Thus, these findings reveal an unexpected general repertoire overlap between MHC class II epitopes recognized in both species of macaques and in humans, showing that epitope pools defined in humans can also be used to characterize macaque responses, despite differences in species and antigen exposure. The results have general implications for the evaluation of new vaccines and diagnostics in NHPs, and immediate applicability in the setting of macaque models of TB.
Collapse
Affiliation(s)
- Bianca R Mothé
- Department of Biology, CSUSM, San Marcos, CA 92096, USA; La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA.
| | | | - Courtney Dow
- Department of Biology, CSUSM, San Marcos, CA 92096, USA
| | - Myles B C Dillon
- La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Roger W Wiseman
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Patrick Bohn
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Julie Karl
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Nadia A Golden
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Trey Gilpin
- Department of Biology, CSUSM, San Marcos, CA 92096, USA
| | - Taylor W Foreman
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15216, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University Baton Rouge, LA 70803, USA
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Department of Pediatrics and Child Health, University of Cape Town, Cape Town 7925, South Africa
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15216, USA
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - David H O'Connor
- Wisconsin National Primate Research Center and Department of Pathology and Laboratory Medicine, UW-Madison, Madison, WI 53706, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| |
Collapse
|
30
|
Gautam US, Mehra S, Kaushal D. In-Vivo Gene Signatures of Mycobacterium tuberculosis in C3HeB/FeJ Mice. PLoS One 2015; 10:e0135208. [PMID: 26270051 PMCID: PMC4535907 DOI: 10.1371/journal.pone.0135208] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/19/2015] [Indexed: 11/28/2022] Open
Abstract
Despite considerable progress in understanding the pathogenesis of Mycobacterium tuberculosis (Mtb), development of new therapeutics and vaccines against it has proven difficult. This is at least in part due to the use of less than optimal models of in-vivo Mtb infection, which has precluded a study of the physiology of the pathogen in niches where it actually persists. C3HeB/FeJ (Kramnik) mice develop human-like lesions when experimentally infected with Mtb and thus make available, a faithful and highly tractable system to study the physiology of the pathogen in-vivo. We compared the transcriptomics of Mtb and various mutants in the DosR (DevR) regulon derived from Kramnik mouse granulomas to those cultured in-vitro. We recently showed that mutant ΔdosS is attenuated in C3HeB/FeJ mice. Aerosol exposure of mice with the mutant mycobacteria resulted in a substantially different and a relatively weaker transcriptional response (< = 20 genes were induced) for the functional category ‘Information Pathways’ in Mtb:ΔdosR; ‘Lipid Metabolism’ in Mtb:ΔdosT; ‘Virulence, Detoxification, Adaptation’ in both Mtb:ΔdosR and Mtb:ΔdosT; and ‘PE/PPE’ family in all mutant strains compare to wild-type Mtb H37Rv, suggesting that the inability to induce DosR functions to different levels can modulate the interaction of the pathogen with the host. The Mtb genes expressed during growth in C3HeB/FeJ mice appear to reflect adaptation to differential nutrient utilization for survival in mouse lungs. The genes such as glnB, Rv0744c, Rv3281, sdhD/B, mce4A, dctA etc. downregulated in mutant ΔdosS indicate their requirement for bacterial growth and flow of carbon/energy source from host cells. We conclude that genes expressed in Mtb during in-vivo chronic phase of infection in Kramnik mice mainly contribute to growth, cell wall processes, lipid metabolism, and virulence.
Collapse
Affiliation(s)
- Uma Shankar Gautam
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- * E-mail: (DK); (USG)
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Louisiana State University School of Veterinary Medicine Department of Pathobiological Sciences, Baton Rouge, Louisiana, United States of America
| | - Deepak Kaushal
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail: (DK); (USG)
| |
Collapse
|
31
|
Devasundaram S, Khan I, Kumar N, Das S, Raja A. The influence of reduced oxygen availability on gene expression in laboratory (H37Rv) and clinical strains (S7 and S10) of Mycobacterium tuberculosis. J Biotechnol 2015; 210:70-80. [PMID: 26001906 DOI: 10.1016/j.jbiotec.2015.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/16/2015] [Accepted: 04/23/2015] [Indexed: 11/17/2022]
Abstract
Mycobacterium tuberculosis has the ability to persist within the host in a dormant stage. One important condition believed to contribute to dormancy is reduced access to oxygen known as hypoxia. However, the response of M. tuberculosis to such hypoxia condition is not fully characterized. Virtually all dormant models against tuberculosis tested in animals used laboratory strain H37Rv or Erdman strain. But major outbreaks of tuberculosis (TB) occur with the strains that have widely different genotypes and phenotypes compared to H37Rv. In this study, we used a custom oligonucleotide microarray to determine the overall transcriptional response of laboratory strain (H37Rv) and most prevalent clinical strains (S7 and S10) of M. tuberculosis from South India to hypoxia. Analysis of microarray results revealed that a total of 1161 genes were differentially regulated (≥1.5 fold change) in H37Rv, among them 659 genes upregulated and 502 genes down regulated. Microarray data of clinical isolates showed that a total of 790 genes were differentially regulated in S7 among which 453 genes were upregulated and 337 down regulated. Interestingly, numerous genes were also differentially regulated in S10 (total 2805 genes) of which 1463 genes upregulated and 1342 genes down regulated during reduced oxygen condition (Wayne's model). One hundred and thirty-four genes were found common and upregulated among all three strains (H37Rv, S7, and S10) and can be targeted for drug/vaccine development against TB.
Collapse
Affiliation(s)
- Santhi Devasundaram
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), (Formerly Tuberculosis Research Centre), No. 1, Mayor Sathiyamoorthy Road, Chetpet, Chennai 600 031, India
| | - Imran Khan
- Department of Molecular Reproduction, Development and Genetics Biological Sciences Building, Indian Institute of Science, Bangalore 560 012, India
| | - Neeraj Kumar
- Department of Molecular Reproduction, Development and Genetics Biological Sciences Building, Indian Institute of Science, Bangalore 560 012, India
| | - Sulochana Das
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), (Formerly Tuberculosis Research Centre), No. 1, Mayor Sathiyamoorthy Road, Chetpet, Chennai 600 031, India
| | - Alamelu Raja
- Department of Immunology, National Institute for Research in Tuberculosis (ICMR), (Formerly Tuberculosis Research Centre), No. 1, Mayor Sathiyamoorthy Road, Chetpet, Chennai 600 031, India.
| |
Collapse
|
32
|
Narrandes NC, Machowski EE, Mizrahi V, Kana BD. Cleavage of the moaX-encoded fused molybdopterin synthase from Mycobacterium tuberculosis is necessary for activity. BMC Microbiol 2015; 15:22. [PMID: 25651977 PMCID: PMC4326299 DOI: 10.1186/s12866-015-0355-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/19/2015] [Indexed: 11/21/2022] Open
Abstract
Background Molybdopterin cofactor (MoCo) biosynthesis in Mycobacterium tuberculosis is associated with a multiplicity of genes encoding several enzymes in the pathway, including the molybdopterin (MPT) synthase, a hetero tetramer comprising two MoaD and two MoaE subunits. In addition to moaD1, moaD2, moaE1, moaE2, the M. tuberculosis genome also contains a moaX gene which encodes an MPT-synthase in which the MoaD and MoaE domains are located on a single polypeptide. In this study, we assessed the requirement for post-translational cleavage of MoaX for functionality of this novel, fused MPT synthase and attempted to establish a functional hierarchy for the various MPT-synthase encoding genes in M. tuberculosis. Results Using a heterologous Mycobacterium smegmatis host and the activity of the MoCo-dependent nitrate reductase, we confirmed that moaD2 and moaE2 from M. tuberculosis together encode a functional MPT synthase. In contrast, moaD1 displayed no functionality in this system, even in the presence of the MoeBR sulphurtransferase, which contains the rhodansese-like domain, predicted to activate MoaD subunits. We demonstrated that cleavage of MoaX into its constituent MoaD and MoaE subunits was required for MPT synthase activity and confirmed that cleavage occurs between the Gly82 and Ser83 residues in MoaX. Further analysis of the Gly81-Gly82 motif confirmed that both of these residues are necessary for catalysis and that the Gly81 was required for recognition/cleavage of MoaX by an as yet unidentified protease. In addition, the MoaE component of MoaX was able to function in conjunction with M. smegmatis MoaD2 suggesting that cleavage of MoaX renders functionally interchangeable subunits. Expression of MoaX in E. coli revealed that incorrect post-translational processing is responsible for the lack of activity of MoaX in this heterologous host. Conclusions There is a degree of functional interchangeability between the MPT synthase subunits of M. tuberculosis. In the case of MoaX, post-translational cleavage at the Gly82 residue is required for function. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0355-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicole C Narrandes
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, P.O. Box 1038, Johannesburg, 2000, South Africa.
| | - Edith Erika Machowski
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, P.O. Box 1038, Johannesburg, 2000, South Africa.
| | - Valerie Mizrahi
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit and DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease & Molecular Medicine and Division of Medical Microbiology, University of Cape Town, Cape Town, South Africa.
| | - Bavesh D Kana
- DST/NRF Centre of Excellence for Biomedical TB Research, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Service, P.O. Box 1038, Johannesburg, 2000, South Africa.
| |
Collapse
|
33
|
Phillips BL, Mehra S, Ahsan MH, Selman M, Khader SA, Kaushal D. LAG3 expression in active Mycobacterium tuberculosis infections. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:820-33. [PMID: 25549835 DOI: 10.1016/j.ajpath.2014.11.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 10/22/2014] [Accepted: 11/03/2014] [Indexed: 01/22/2023]
Abstract
Mycobacterium tuberculosis (MTB) is a highly successful pathogen because of its ability to persist in human lungs for long periods of time. MTB modulates several aspects of the host immune response. Lymphocyte-activation gene 3 (LAG3) is a protein with a high affinity for the CD4 receptor and is expressed mainly by regulatory T cells with immunomodulatory functions. To understand the function of LAG3 during MTB infection, a nonhuman primate model of tuberculosis, which recapitulates key aspects of natural human infection in rhesus macaques (Macaca mulatta), was used. We show that the expression of LAG3 is highly induced in the lungs and particularly in the granulomatous lesions of macaques experimentally infected with MTB. Furthermore, we show that LAG3 expression is not induced in the lungs and lung granulomas of animals exhibiting latent tuberculosis infection. However, simian immunodeficiency virus-induced reactivation of latent tuberculosis infection results in an increased expression of LAG3 in the lungs. This response is not observed in nonhuman primates infected with non-MTB bacterial pathogens, nor with simian immunodeficiency virus alone. Our data show that LAG3 was expressed primarily on CD4(+) T cells, presumably by regulatory T cells but also by natural killer cells. The expression of LAG3 coincides with high bacterial burdens and changes in the host type 1 helper T-cell response.
Collapse
Affiliation(s)
- Bonnie L Phillips
- Division of Bacteriology, Tulane National Primate Research Center, Covington, Louisiana; Biomedical Sciences Graduate Student Program, New Orleans, Louisiana; National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Smriti Mehra
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana
| | - Muhammad H Ahsan
- Division of Bacteriology, Tulane National Primate Research Center, Covington, Louisiana; Training in Lung Molecular and Cell Pathobiology Program, New Orleans, Louisiana
| | - Moises Selman
- National Institute of Respiratory Diseases, Mexico City, Mexico
| | - Shabaana A Khader
- Department of Molecular Microbiology and Immunology, Washington University of St. Louis, St. Louis, Missouri
| | - Deepak Kaushal
- Division of Bacteriology, Tulane National Primate Research Center, Covington, Louisiana; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
34
|
McGillivray A, Golden NA, Kaushal D. The Mycobacterium tuberculosis Clp gene regulator is required for in vitro reactivation from hypoxia-induced dormancy. J Biol Chem 2014; 290:2351-67. [PMID: 25422323 DOI: 10.1074/jbc.m114.615534] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the leading cause of death from an infectious disease worldwide and is the causative agent of tuberculosis (Chao, M. C., and Rubin, E. J. (2010) Annu. Rev. Microbiol. 64, 293-311). Throughout infection, Mtb encounters a variety of host pressures. Thus, responding to these host stresses via the induction of multiple regulatory networks is needed for survival within the host. The Clp protease gene regulator, Rv2745c (clgR), is induced in response to environmental stress conditions, implicating its potential role in Mtb pathogenesis. Transcriptional activation of genes downstream of Rv2745c occurs in a condition-dependent manner. Our isogenic Mtb:ΔRv2745c mutant expresses a significantly different phenotype upon reaeration conditions. Transcriptional analysis revealed differential gene expression profiles relative to wild-type Mtb. Rv2745c is strongly induced in response to hypoxic and reaeration conditions, implicating a role of Rv2745c in vivo during both establishment of infection and reactivation. We found dysregulation of downstream genes within both the σ(H)/σ(E) regulon as well as the dosR regulon in the isogenic mutant, Mtb:ΔRv2745c. Upon hypoxic and reaeration conditions, Clp protease induction occurred within wild-type Mtb, indicating that activation of clgR, which subsequently leads to Clp protease induction, is crucial for degradation of misfolded proteins and ultimately survival of Mtb upon specific stress conditions. Our data indicate the diverse response of Rv2745c, σ(H) and σ(E) in response to a variety of stress conditions. Activation of Rv2745c in response to various stress conditions leads to differential activation of downstream genes, indicating the diverse role of Rv2745c and its importance for Mtb survival in vivo.
Collapse
Affiliation(s)
- Amanda McGillivray
- From the Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana 70433
| | - Nadia A Golden
- From the Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana 70433
| | - Deepak Kaushal
- From the Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana 70433
| |
Collapse
|
35
|
Prozorov AA, Fedorova IA, Bekker OB, Danilenko VN. The virulence factors of Mycobacterium tuberculosis: Genetic control, new conceptions. RUSS J GENET+ 2014. [DOI: 10.1134/s1022795414080055] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
36
|
Li D, Zhang C, Lu N, Mu L, He Y, Xu L, Yang J, Fan Y, Kang Y, Yang C. Cloning and characterization of Clp protease proteolytic subunit 2 and its implication in clinical diagnosis of tuberculosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:5674-5682. [PMID: 25337208 PMCID: PMC4203179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/20/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVE To clone, express, and characterize Mycobacterium tuberculosis (Mtb) ClpP2, and evaluated the potential usage of ClpP2 in clinical diagnosis of tuberculosis. METHODS Mtb ClpP2 was cloned into recombinant plasmid pET32a (+) and transformed into E. coli BL21 (DE3). SDS-PAGE and Western blot analysis were performed to detect the expression of the recombinant protein. The immunogenicity of Mtb ClpP2 was assessed with epitope prediction and antibody titer assay. Quantitative real-time PCR was performed to detect the influence of stress conditions on ClpP2 expression. ClpP2 antigen and antibody in patients with pulmonary diseases were detected by indirect ELISA. ROC curve was constructed to assess the diagnostic accuracy of Mtb ClpP2 for tuberculosis. RESULTS We had cloned and expressed recombinant Mtb ClpP2 in E. coli. Our results showed that Mtb ClpP2 had potent immunogenicity, and our own prepared polyclonal antibody could be used in detection and diagnostic tests. Results from Western blot showed that ClpP2 was mainly located in M. bovis BCG cytoplasm, and real-time PCR indicated that stress conditions could enhance the mRNA expression of ClpP2. Indirect ELISA suggested that, in tuberculosis patients, both the levels of ClpP2 antigen and antibody were increased, and the positive rates of ClpP2 were elevated. ROC curve had demonstrated satisfactory sensitivity and specificity of ClpP2-based diagnosis for tuberculosis. CONCLUSION Our results suggest that Mtb ClpP2 antigens would be used as a biomarker in tuberculosis pathogenesis. These findings highlight the feasibility of the application of Mtb ClpP2 in the clinical diagnosis of tuberculosis.
Collapse
Affiliation(s)
- Dairong Li
- Department of Respiratory Disease, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
- Department of Pathology, Chongqing Medical UniversityChongqing 400016, China
| | - Chunyan Zhang
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Nan Lu
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Liuqing Mu
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Yonglin He
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Lei Xu
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Jing Yang
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Yu Fan
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Yuexi Kang
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| | - Chun Yang
- Department of Pathogenic Organisms, Chongqing Medical UniversityChongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
37
|
Parandhaman DK, Sharma P, Bisht D, Narayanan S. Proteome and phosphoproteome analysis of the serine/threonine protein kinase E mutant of Mycobacterium tuberculosis. Life Sci 2014; 109:116-26. [PMID: 24972353 DOI: 10.1016/j.lfs.2014.06.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/23/2014] [Accepted: 06/09/2014] [Indexed: 01/04/2023]
Abstract
AIMS Serine/threonine protein kinases (STPKs) have prominent roles in the survival mechanisms of Mycobacterium tuberculosis (M. tuberculosis). Previous studies from our laboratory underscored the role of PknE, an STPK in virulence, adaptation and the suppression of host cell apoptosis. In this study, two-dimensional gel electrophoresis was used to study the proteome and phosphoproteome profiles of wild type M. tuberculosis and its isogenic pknE deletion mutant (ΔpknE) during growth in Middlebrook 7H9 and nitric oxide stress. MAIN METHODS Wild-type M. tuberculosis and its isogenic pknE deletion mutant strain were grown in Middlebrook 7H9 as well as subjected to nitric oxide stress using sodium nitroprusside. Whole cell lysates were prepared and analyzed by 2D-gel electrophoresis. Phosphoproteomes were analyzed using phospho serine and phospho threonine antibodies after subjecting the 2D-gels to western blotting. Proteins of interest were identified using mass spectrometry. KEY FINDINGS Our analysis provides insights into the targets that impose pro-apoptotic as well as altered cellular phenotypes on ΔpknE, revealing novel substrates and functions for PknE. SIGNIFICANCE For the first time, our proteome and phosphoproteome data decipher the function of PknE in cell division, virulence, dormancy, suppression of sigma factor B and its regulated genes, suppression of two-component systems and in the metabolic activity of M. tuberculosis.
Collapse
Affiliation(s)
- Dinesh Kumar Parandhaman
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai,India; Department of Immunology, International Centre for Genetic Engineering and Biotechnology, Aruna Asif Ali Marg, New Delhi, 110067,India
| | - Prashant Sharma
- Department of Biochemistry, National JALMA Institute for Leprosy and other Mycobacterial Diseases, Tajganj, Agra,India
| | - Deepa Bisht
- Department of Biochemistry, National JALMA Institute for Leprosy and other Mycobacterial Diseases, Tajganj, Agra,India
| | - Sujatha Narayanan
- Department of Immunology, National Institute for Research in Tuberculosis, Chennai,India.
| |
Collapse
|
38
|
Hudock TA, Kaushal D. A novel microdissection approach to recovering mycobacterium tuberculosis specific transcripts from formalin fixed paraffin embedded lung granulomas. J Vis Exp 2014. [PMID: 24962586 DOI: 10.3791/51693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Microdissection has been used for the examination of tissues at DNA, RNA, and protein levels for over a decade. Laser capture microscopy (LCM) is the most common microdissection technique used today. In this technique, a laser is used to focally melt a thermoplastic membrane that overlies a dehydrated tissue section(1). The tissue section composite is then lifted and separated from the membrane. Although this technique can be used successfully for tissue examination, it is time consuming and expensive. Furthermore, the successful completion of procedures using this technique requires the use of a laser, thus limiting its use. A new more affordable and practical microdissection approach called mesodissection is a possible solution to the pitfalls of LCM. This technique employs the MESO-1/MeSectr system to mill the desired tissue from a slide mounted tissue sample while concurrently dispensing and aspirating fluid to recover the desired tissue sample into a consumable mill bit. Before the dissection process begins, the user aligns the formalin fixed paraffin embedded (FFPE) slide with a hematoxylin and eosin stained (H&E) reference slide. Thereafter, the operator annotates the desired dissection area and proceeds to dissect the appropriate segment. The program generates an archived image of the dissection. The main advantage of mesodissection is the short duration needed to dissect a slide, taking an average of ten minutes from set up to sample generation in this experiment. Additionally, the system is significantly more cost effective and user friendly. A slight disadvantage is that it is not as precise as laser capture microscopy. In this article we demonstrate how mesodissection can be used to extract RNA from slides from FFPE granulomas caused by Mycobacterium tuberculosis (Mtb).
Collapse
Affiliation(s)
- Teresa A Hudock
- Bacteriology and Parasitology, Tulane National Primate Research Center;
| | - Deepak Kaushal
- Bacteriology and Parasitology, Tulane National Primate Research Center; Microbiology and Immunology, Tulane National Primate Research Center
| |
Collapse
|
39
|
Wei J, Liang J, Shi Q, Yuan P, Meng R, Tang X, Yu L, Guo N. Genome-wide transcription analyses in Mycobacterium tuberculosis treated with lupulone. Braz J Microbiol 2014; 45:333-41. [PMID: 24948953 PMCID: PMC4059319 DOI: 10.1590/s1517-83822014005000032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 04/01/2013] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis, still causes higher mortality than any other bacterial pathogen until now. With the emergence and spread of multidrug-resistant (MDR) and extensively drug-resistant (XDR-TB) strains, it becomes more important to search for alternative targets to develop new antimycobacterial drugs. Lupulone is a compound extracted from Hops (Hurnulus lupulus), which exhibits a good antimicrobial activity against M. tuberculosis with minimal inhibitory concentration (MIC) value of 10 μg/mL, but the response mechanisms of lupulone against M. tuberculosis are still poorly understood. In this study, we used a commercial oligonucleotide microarray to determine the overall transcriptional response of M. tuberculosis H37Rv triggered by exposure to MIC of lupulone. A total of 540 genes were found to be differentially regulated by lupulone. Of these, 254 genes were upregulated, and 286 genes were downregulated. A number of important genes were significantly regulated which are involved in various pathways, such as surface-exposed lipids, cytochrome P450 enzymes, PE/PPE multigene families, ABC transporters, and protein synthesis. Real-time quantitative RT-PCR was performed for choosed genes to verified the microarray results. To our knowledge, this genome-wide transcriptomics approach has produced the first insights into the response of M. tuberculosis to a lupulone challenge.
Collapse
Affiliation(s)
- Jian Wei
- Life Science Department ChangChun Normal University Changchun P.R. China ; Bio-Reactor Center Jilin Agrichultural University Changchun P.R. China
| | - Junchao Liang
- Department of Food Quality and Safety Key Laboratory of Zoonosis Research Institute of Zoonosis Ministry of Education Changchun P. R. China
| | - Qiyun Shi
- Department of Food Quality and Safety Key Laboratory of Zoonosis Research Institute of Zoonosis Ministry of Education Changchun P. R. China
| | - Peng Yuan
- Department of Food Quality and Safety Key Laboratory of Zoonosis Research Institute of Zoonosis Ministry of Education Changchun P. R. China
| | - Rizeng Meng
- Jilin Enrty-Exit Inspection and Quarantine Bureau Changchun P.R. China
| | - Xudong Tang
- Key Lab for New Drug Research of TCM Research Institute of Tsinghua University in Shenzhen Shenzhen P.R. China
| | - Lu Yu
- Department of Food Quality and Safety Key Laboratory of Zoonosis Research Institute of Zoonosis Ministry of Education Changchun P. R. China
| | - Na Guo
- Department of Food Quality and Safety Key Laboratory of Zoonosis Research Institute of Zoonosis Ministry of Education Changchun P. R. China
| |
Collapse
|
40
|
McGillivray A, Golden NA, Gautam US, Mehra S, Kaushal D. The Mycobacterium tuberculosis Rv2745c plays an important role in responding to redox stress. PLoS One 2014; 9:e93604. [PMID: 24705585 PMCID: PMC3976341 DOI: 10.1371/journal.pone.0093604] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/07/2014] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is the leading cause of death from an infectious disease worldwide. Over the course of its life cycle in vivo, Mtb is exposed to a plethora of environmental stress conditions. Temporal regulation of genes involved in sensing and responding to such conditions is therefore crucial for Mtb to establish an infection. The Rv2745c (clgR) gene encodes a Clp protease gene regulator that is induced in response to a variety of stress conditions and potentially plays a role in Mtb pathogenesis. Our isogenic mutant, Mtb:ΔRv2745c, is significantly more sensitive to in vitro redox stress generated by diamide, relative to wild-type Mtb as well as to a complemented strain. Together with the fact that the expression of Rv2745c is strongly induced in response to redox stress, these results strongly implicate a role for ClgR in the management of intraphagosomal redox stress. Additionally, we observed that redox stress led to the dysregulation of the expression of the σH/σE regulon in the isogenic mutant, Mtb:ΔRv2745c. Furthermore, induction of clgR in Mtb and Mtb:ΔRv2745c (comp) did not lead to Clp protease induction, indicating that clgR has additional functions that need to be elucidated. Our data, when taken together with that obtained by other groups, indicates that ClgR plays diverse roles in multiple regulatory networks in response to different stress conditions. In addition to redox stress, the expression of Rv2745c correlates with the expression of genes involved in sulfate assimilation as well as in response to hypoxia and reaeration. Clearly, the Mtb Rv2745c-encoded ClgR performs different functions during stress response and is important for the pathogenicity of Mtb in-vivo, regardless of its induction of the Clp proteolytic pathway.
Collapse
Affiliation(s)
- Amanda McGillivray
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Nadia Abrahams Golden
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Uma Shankar Gautam
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Smriti Mehra
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Deepak Kaushal
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
41
|
Hatzios SK, Baer CE, Rustad TR, Siegrist MS, Pang JM, Ortega C, Alber T, Grundner C, Sherman DR, Bertozzi CR. Osmosensory signaling in Mycobacterium tuberculosis mediated by a eukaryotic-like Ser/Thr protein kinase. Proc Natl Acad Sci U S A 2013; 110:E5069-77. [PMID: 24309377 PMCID: PMC3876250 DOI: 10.1073/pnas.1321205110] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Bacteria are able to adapt to dramatically different microenvironments, but in many organisms, the signaling pathways, transcriptional programs, and downstream physiological changes involved in adaptation are not well-understood. Here, we discovered that osmotic stress stimulates a signaling network in Mycobacterium tuberculosis regulated by the eukaryotic-like receptor Ser/Thr protein kinase PknD. Expression of the PknD substrate Rv0516c was highly induced by osmotic stress. Furthermore, Rv0516c disruption modified peptidoglycan thickness, enhanced antibiotic resistance, and activated genes in the regulon of the alternative σ-factor SigF. Phosphorylation of Rv0516c regulated the abundance of EspA, a virulence-associated substrate of the type VII ESX-1 secretion system. These findings identify an osmosensory pathway orchestrated by PknD, Rv0516c, and SigF that enables adaptation to osmotic stress through cell wall remodeling and virulence factor production. Given the widespread occurrence of eukaryotic-like Ser/Thr protein kinases in bacteria, these proteins may play a broad role in bacterial osmosensing.
Collapse
Affiliation(s)
| | - Christina E. Baer
- Molecular and Cell Biology, California Institute for Quantitative Biosciences (QB3), and
| | - Tige R. Rustad
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
- Department of Global Health, University of Washington, Seattle, WA 98195
| | | | - Jennifer M. Pang
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
- Department of Global Health, University of Washington, Seattle, WA 98195
| | - Corrie Ortega
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
- Department of Global Health, University of Washington, Seattle, WA 98195
| | - Tom Alber
- Molecular and Cell Biology, California Institute for Quantitative Biosciences (QB3), and
| | - Christoph Grundner
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
- Department of Global Health, University of Washington, Seattle, WA 98195
| | - David R. Sherman
- Seattle Biomedical Research Institute, Seattle, WA 98109; and
- Department of Global Health, University of Washington, Seattle, WA 98195
| | - Carolyn R. Bertozzi
- Departments of Chemistry and
- Molecular and Cell Biology, California Institute for Quantitative Biosciences (QB3), and
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720
| |
Collapse
|
42
|
Joung SM, Ryoo S. BCG vaccine in Korea. Clin Exp Vaccine Res 2013; 2:83-91. [PMID: 23858398 PMCID: PMC3710928 DOI: 10.7774/cevr.2013.2.2.83] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/10/2013] [Accepted: 03/20/2013] [Indexed: 11/30/2022] Open
Abstract
The anti-tuberculosis Bacille de Calmette et Guérin (BCG) vaccine was developed between 1905 and 1921 at Pasteur Institutes of Lille in France, and was adopted by many countries. BCG strains comprise natural mutants of major virulence factors of Mycobacterium tuberculosis and that BCG sub-strains differ markedly in virulence levels. The tuberculosis became endemic in Korea after the Korean War (1950s). The BCG strain, which was donated by Pasteur Institutes, was brought to Korea in 1955, and the first domestic BCG vaccine was produced by the National Defense Research Institute (NDRI), current Korea Centers for Disease Control and Prevention (KCDC), in 1960. Since 1987, BCG manufacture work was handed over to the Korean Institute of Tuberculosis (KIT), the freeze-dried BCG vaccine was manufactured at a scale required to meet the whole amount of domestic consumption. However, since 2006, the manufacture of BCG vaccine suspended and the whole amount of BCG was imported at this point of time. Now KIT is planning to re-produce the BCG vaccine in Korea under the supervision of KCDC, this will be render great role to National Tuberculosis Control Program (NTP) and provide initiating step for developing new tuberculosis vaccines in Korea.
Collapse
Affiliation(s)
- Sun Myung Joung
- Korean Institute of Tuberculosis, The Korean National Tuberculosis Association, Cheongwon, Korea
| | | |
Collapse
|
43
|
Key role for the alternative sigma factor, SigH, in the intracellular life of Mycobacterium avium subsp. paratuberculosis during macrophage stress. Infect Immun 2013; 81:2242-57. [PMID: 23569115 DOI: 10.1128/iai.01273-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium avium subsp. paratuberculosis causes Johne's disease, an enteric infection in cattle and other ruminants, greatly afflicting the dairy industry worldwide. Once inside the cell, M. avium subsp. paratuberculosis is known to survive harsh microenvironments, especially those inside activated macrophages. To improve our understanding of M. avium subsp. paratuberculosis pathogenesis, we examined phagosome maturation associated with transcriptional responses of M. avium subsp. paratuberculosis during macrophage infection. Monitoring cellular markers, only live M. avium subsp. paratuberculosis bacilli were able to prevent phagosome maturation and reduce its acidification. On the transcriptional level, over 300 M. avium subsp. paratuberculosis genes were significantly and differentially regulated in both naive and IFN-γ-activated macrophages. These genes include the sigma factor H (sigH) that was shown to be important for M. avium subsp. paratuberculosis survival inside gamma interferon (IFN-γ)-activated bovine macrophages. Interestingly, an sigH-knockout mutant showed increased sensitivity to a sustained level of thiol-specific oxidative stress. Large-scale RNA sequence analysis revealed that a large number of genes belong to the sigH regulon, especially following diamide stress. Genes involved in oxidative stress and virulence were among the induced genes in the sigH regulon with a putative consensus sequence for SigH binding that was recognized in a subset of these genes (n = 30), suggesting direct regulation by SigH. Finally, mice infections showed a significant attenuation of the ΔsigH mutant compared to its parental strain, suggesting a role for sigH in M. avium subsp. paratuberculosis virulence. Such analysis could identify potential targets for further testing as vaccine candidates against Johne's disease.
Collapse
|
44
|
Personne Y, Brown AC, Schuessler DL, Parish T. Mycobacterium tuberculosis ClpP proteases are co-transcribed but exhibit different substrate specificities. PLoS One 2013; 8:e60228. [PMID: 23560081 PMCID: PMC3613350 DOI: 10.1371/journal.pone.0060228] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/23/2013] [Indexed: 11/19/2022] Open
Abstract
Caseinolytic (Clp) proteases are widespread energy-dependent proteases; the functional ATP-dependent protease is comprised of multimers of proteolytic and regulatory subunits. Mycobacterium tuberculosis has two ClpP proteolytic subunits (ClpP1 and ClpP2), with both being essential for growth in vitro. ClpP1 and clpP2 are arranged in an apparent operon; we demonstrated that the two genes are co-expressed under normal growth conditions. We identified a single promoter region for the clpP1P2 operon; no promoter was detected upstream of clpP2 demonstrating that independent expression of clpP1 and clpP2 was highly unlikely. Promoter activity was not induced by heat shock or oxidative stress. We identified a regulatory region upstream of the promoter with a consensus sequence matching the ClgR regulator motif; we determined the limits of the region by mutagenesis and confirmed that positive regulation of the promoter occurs in M. tuberculosis. We developed a reporter system to monitor ClpP1 and ClpP2 enzymatic activities based on LacZ incorporating ssrAtag sequences. We showed that whilst both ClpP1 and ClpP2 degrade SsrA-tagged LacZ, ClpP2 (but not ClpP1) degrades untagged proteins. Our data suggest that the two proteolytic subunits display different substrate specificities and therefore have different, but overlapping roles in M. tuberculosis.
Collapse
Affiliation(s)
- Yoann Personne
- Queen Mary University of London, Barts & The London School of Medicine and Dentistry, London E1 2AT, United Kingdom
| | - Amanda C. Brown
- Queen Mary University of London, Barts & The London School of Medicine and Dentistry, London E1 2AT, United Kingdom
| | - Dorothée L. Schuessler
- Queen Mary University of London, Barts & The London School of Medicine and Dentistry, London E1 2AT, United Kingdom
| | - Tanya Parish
- Queen Mary University of London, Barts & The London School of Medicine and Dentistry, London E1 2AT, United Kingdom
| |
Collapse
|
45
|
Paritala H, Carroll KS. New targets and inhibitors of mycobacterial sulfur metabolism. Infect Disord Drug Targets 2013; 13:85-115. [PMID: 23808874 PMCID: PMC4332622 DOI: 10.2174/18715265113139990022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 05/08/2013] [Indexed: 11/22/2022]
Abstract
The identification of new antibacterial targets is urgently needed to address multidrug resistant and latent tuberculosis infection. Sulfur metabolic pathways are essential for survival and the expression of virulence in many pathogenic bacteria, including Mycobacterium tuberculosis. In addition, microbial sulfur metabolic pathways are largely absent in humans and therefore, represent unique targets for therapeutic intervention. In this review, we summarize our current understanding of the enzymes associated with the production of sulfated and reduced sulfur-containing metabolites in Mycobacteria. Small molecule inhibitors of these catalysts represent valuable chemical tools that can be used to investigate the role of sulfur metabolism throughout the Mycobacterial lifecycle and may also represent new leads for drug development. In this light, we also summarize recent progress made in the development of inhibitors of sulfur metabolism enzymes.
Collapse
Affiliation(s)
| | - Kate S. Carroll
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida, 33458, USA
| |
Collapse
|
46
|
Nambu T, Yamane K, Yamanaka T, Mashimo C, Maruyama H, Yoshida M, Hayashi H, Leung KP, Fukushima H. Identification of disulphide stress-responsive extracytoplasmic function sigma factors in Rothia mucilaginosa. Arch Oral Biol 2013; 58:681-9. [PMID: 23399044 DOI: 10.1016/j.archoralbio.2012.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/21/2012] [Accepted: 10/23/2012] [Indexed: 11/17/2022]
Abstract
Rothia mucilaginosa is known as a member of commensal bacterial flora in the oral cavity and has received attention as a potential opportunistic pathogen. We previously determined the genomic sequence of R. mucilaginosa DY-18, a clinical strain with biofilm-like structures isolated from an infected root canal of a tooth with persistent apical periodontitis. We found that the DY-18 genome had only two sigma factor genes that encoded the primary and extracytoplasmic function (ECF) sigma factors. Genomic analysis on the available database of R. mucilaginosa ATCC 25296 (a type strain for R. mucilaginosa) revealed that ATCC 25296 has three sigma factors: one primary sigma factor and two ECF sigma factors, one of which was highly homologous to that of DY-18. ECF sigma factors play an important role in the response to environmental stress and to the production of virulence factors. Therefore, we first examined gene-encoding sigma factors on R. mucilaginosa genome in silico. The homologous ECF sigma factors found in strains DY-18 and ATCC 25296 formed a distinct SigH (SigR) clade in a phylogenetic tree and their cognate anti-sigma factor has a HXXXCXXC motif known to respond against disulphide stress. Quantitative reverse transcription polymerase chain reaction (PCR) and microarray analysis showed that the transcriptional levels of sigH were markedly up-regulated under disulphide stress in both strains. Microarray data also demonstrated that several oxidative-stress-related genes (thioredoxin, mycothione reductase, reductase and oxidoreductase) were significantly up-regulated under the diamide stress. On the basis of these results, we conclude that the alternative sigma factor SigH of R. mucilaginosa is a candidate regulator in the redox state.
Collapse
Affiliation(s)
- Takayuki Nambu
- Department of Bacteriology, Osaka Dental University, 8-1 Kuzuha-Hanazono, Hirakata, 573-1121 Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Martinez AN, Mehra S, Kaushal D. Role of interleukin 6 in innate immunity to Mycobacterium tuberculosis infection. J Infect Dis 2013; 207:1253-61. [PMID: 23359591 DOI: 10.1093/infdis/jit037] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Mycobacterium tuberculosis can grow in the hostile intracellular environment of macrophages by actively evading macrophage-associated antibacterial activities. The stress response factor SigH contributes to this process by modulating β-chemokine and interleukin 6 (Il6) expression. Hence, Il6 is of critical importance for acquired immunity against M. tuberculosis infection. Here, we attempted to better characterize the role of Il6 in the immune response to M. tuberculosis infection. METHODS A small interfering RNA-based approach was used to silence expression of the Il6 transcript in host macrophages infected with a wild-type strain of M. tuberculosis or an attenuated mutant strain of M. tuberculosis (Mtb:Δ-sigH). The outcome was measured by the analysis of bacterial burden and transcriptome-wide analysis of host gene expression. Transcriptome results were confirmed via quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS Wild type and Mtb:Δ-sigH infection of host macrophages in which Il6 had been silenced resulted in increased expression of interferon-inducible genes, especially those involved in type I interferon signaling. The expression of Ly-6 genes was significantly higher in cells infected with Mtb:Δ-sigH, compared with those infected with the wild-type strain (P < .05). CONCLUSIONS M. tuberculosis regulates host Il6 production to inhibit type I interferon signaling and, consequently, disease progression. Mtb:Δ-sigH is associated with delayed activation of macrophages, compared with the wild-type strain, and with delayed inflammatory stimuli as consequence. These findings have important implications for improving understanding of the mechanisms behind M. tuberculosis virulence and pathogenesis and provide an initial road map to further investigate the mechanisms that may account for the deleterious effects of type I interferons in M. tuberculosis infection.
Collapse
Affiliation(s)
- Alejandra N Martinez
- Divisions of Bacteriology and Parasitology, Tulane National Primate Research Center, Covington, LA 70433, USA
| | | | | |
Collapse
|
48
|
Williams M, Mizrahi V, Kana BD. Molybdenum cofactor: a key component of Mycobacterium tuberculosis pathogenesis? Crit Rev Microbiol 2013; 40:18-29. [PMID: 23317461 DOI: 10.3109/1040841x.2012.749211] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mycobacterium tuberculosis (Mtb) and other members of the Mtb complex possess an expanded complement of genes for the biosynthesis of molybdenum cofactor (MoCo), a tricyclic pterin molecule that is covalently attached to molybdate. This cofactor allows the redox properties of molybdenum to be harnessed by enzymes in order to catalyze redox reactions in carbon, nitrogen and sulfur metabolism. In this article, we summarize recent advances in elucidating the MoCo biosynthetic pathway in Mtb and highlight the evidence implicating the biosynthesis of this cofactor, as well as the enzymes that depend upon it for activity, in Mtb pathogenesis.
Collapse
Affiliation(s)
- Monique Williams
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Division of Medical Microbiology, Faculty of Health Sciences , University of Cape Town
| | | | | |
Collapse
|
49
|
Forrellad MA, Klepp LI, Gioffré A, Sabio y García J, Morbidoni HR, de la Paz Santangelo M, Cataldi AA, Bigi F. Virulence factors of the Mycobacterium tuberculosis complex. Virulence 2012; 4:3-66. [PMID: 23076359 PMCID: PMC3544749 DOI: 10.4161/viru.22329] [Citation(s) in RCA: 406] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Mycobacterium tuberculosis complex (MTBC) consists of closely related species that cause tuberculosis in both humans and animals. This illness, still today, remains to be one of the leading causes of morbidity and mortality throughout the world. The mycobacteria enter the host by air, and, once in the lungs, are phagocytated by macrophages. This may lead to the rapid elimination of the bacillus or to the triggering of an active tuberculosis infection. A large number of different virulence factors have evolved in MTBC members as a response to the host immune reaction. The aim of this review is to describe the bacterial genes/proteins that are essential for the virulence of MTBC species, and that have been demonstrated in an in vivo model of infection. Knowledge of MTBC virulence factors is essential for the development of new vaccines and drugs to help manage the disease toward an increasingly more tuberculosis-free world.
Collapse
|
50
|
Zhang Y, Zheng G, Wang Y, Chen J, Zhu C, Liu R, Peng Z, Li Q, Xing L. Screening and comparison of differentially expressed genes between one MDR-TB strain and the virulent M. tuberculosis H37Rv. Gene 2012; 506:223-9. [DOI: 10.1016/j.gene.2012.06.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/05/2012] [Accepted: 06/17/2012] [Indexed: 10/28/2022]
|