1
|
do Nascimento MEC, Montagna LC, Manfroi L, Laaf YDO, Maiochi LF, da Silva MAC, Lima AODS. Newly Isolated Priestia megaterium LAMA1607 for Enhanced Biological Phosphorus Removal: A Genomic and Functional Characterization. Front Biosci (Elite Ed) 2024; 16:37. [PMID: 39736010 DOI: 10.31083/j.fbe1604037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/21/2024] [Accepted: 09/13/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Enhanced biological phosphorus removal (EBPR) systems utilize phosphorus-accumulating organisms (PAOs) to remove phosphorus from wastewater since excessive phosphorus in water bodies can lead to eutrophication. This study aimed to characterize a newly isolated PAO strain for its potential application in EBPR systems and to screen for additional biotechnological potential. Here, sequencing allowed for genomic analysis, identifying the genes and molecules involved, and exploring other potentials. Additionally, assessing the phosphorus removal performance of the PAO strain in common effluents is essential for its potential application in large-scale systems. METHODS A strain designated LAMA1607 was isolated from activated sludge and selected based on its ability to remove total phosphate from the culture medium. Genomic DNA was extracted and sequenced using the Illumina NovaSeq 6000 platform. Assembly and annotation were performed using CLC Genomics Workbench v.24.0 (QIAGEN®) and Rapid Annotation using Subsystem Technology (RAST)/Pathosystems Resource Integration Center (PATRIC) server tools. Functional prediction of uncharacterized proteins was completed using PHYRE2, and secondary metabolite identification was performed using antiSMASH. Further, additional enzymes with biotechnological applications were manually curated through the Association of Manufacturers and Formulators of Enzyme Products (AMFEP) list. The phosphorus removal capability was assessed in domestic and fishery effluents under enriched and unenriched conditions, where pH, microbial growth, and total phosphorus were monitored over 48 hours. RESULTS The genome sequence comprised 5,234,874 bp divided into 20 contigs, 5540 coding sequences, and a GC content of 38.0%; subsequently, LAMA1607 was identified through Basic Local Alignment Search Tool (BLAST) analysis as Priestia megaterium. Genome annotation revealed 27 genes potentially involved in phosphorus removal, including eight encoding transport proteins, three regulatory proteins, twelve enzymes, and others related to phosphorus incorporation and polyphosphate (polyP) granule formation. Moreover, other enzymes of interest were identified, such as hydrolases, lipases, proteases, and amylases, alongside secondary metabolite gene clusters, such as Non-ribosomal peptide synthetase-independent siderophore. P. megaterium LAMA1607 effectively removed up to 70% of the total phosphorus from the fishery effluent. CONCLUSIONS Genomic analysis suggests that P. megaterium LAMA1607 possesses the mechanistic functions for phosphorus uptake, transport, and storage while also identifying additional biotechnologically relevant enzymes and capabilities. Meanwhile, tests on the effluent demonstrated significant phosphorus removal. These findings support the biotechnological potential and application of P. megaterium LAMA1607 in EBPR systems.
Collapse
Affiliation(s)
| | | | - Laíza Manfroi
- Polytechnic School, University of Vale do Itajaí (Univali), Itajaí, SC 88302-202, Brazil
| | - Yan de Oliveira Laaf
- Polytechnic School, University of Vale do Itajaí (Univali), Itajaí, SC 88302-202, Brazil
| | - Luigi Ferrazza Maiochi
- Polytechnic School, University of Vale do Itajaí (Univali), Itajaí, SC 88302-202, Brazil
| | | | | |
Collapse
|
2
|
Schoeppe R, Waldmann M, Jessen HJ, Renné T. An Update on Polyphosphate In Vivo Activities. Biomolecules 2024; 14:937. [PMID: 39199325 PMCID: PMC11352482 DOI: 10.3390/biom14080937] [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: 06/14/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 09/01/2024] Open
Abstract
Polyphosphate (polyP) is an evolutionary ancient inorganic molecule widespread in biology, exerting a broad range of biological activities. The intracellular polymer serves as an energy storage pool and phosphate/calcium ion reservoir with implications for basal cellular functions. Metabolisms of the polymer are well understood in procaryotes and unicellular eukaryotic cells. However, functions, regulation, and association with disease states of the polymer in higher eukaryotic species such as mammalians are just beginning to emerge. The review summarises our current understanding of polyP metabolism, the polymer's functions, and methods for polyP analysis. In-depth knowledge of the pathways that control polyP turnover will open future perspectives for selective targeting of the polymer.
Collapse
Affiliation(s)
- Robert Schoeppe
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
| | - Henning J. Jessen
- Institute of Organic Chemistry, Albert-Ludwigs-University of Freiburg, D-79105 Freiburg, Germany;
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine (O26), University Medical Center Hamburg-Eppendorf, D-20246 Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
- Center for Thrombosis and Haemostasis (CTH), Johannes Gutenberg University Medical Center, D-55131 Mainz, Germany
| |
Collapse
|
3
|
Liu X, Hu J, Wang W, Yang H, Tao E, Ma Y, Sha S. Mycobacterial Biofilm: Mechanisms, Clinical Problems, and Treatments. Int J Mol Sci 2024; 25:7771. [PMID: 39063012 PMCID: PMC11277187 DOI: 10.3390/ijms25147771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Tuberculosis (TB) remains a threat to human health worldwide. Mycobacterium tuberculosis (Mtb) and other nontuberculous mycobacteria (NTM) can form biofilms, and in vitro and animal experiments have shown that biofilms cause serious drug resistance and mycobacterial persistence. Deeper investigations into the mechanisms of mycobacterial biofilm formation and, consequently, the exploration of appropriate antibiofilm treatments to improve the efficiency of current anti-TB drugs will be useful for curing TB. In this review, the genes and molecules that have been recently reported to be involved in mycobacterial biofilm development, such as ABC transporter, Pks1, PpiB, GroEL1, MprB, (p)ppGpp, poly(P), and c-di-GMP, are summarized. Biofilm-induced clinical problems, including biofilm-related infections and enhanced virulence, as well as their possible mechanisms, are also discussed in detail. Moreover, we also illustrate newly synthesized anti-TB agents that target mycobacterial biofilm, as well as some assistant methods with high efficiency in reducing biofilms in hosts, such as the use of nanoparticles.
Collapse
Affiliation(s)
- Xining Liu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| | - Junxing Hu
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| | - Wenzhen Wang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| | - Hanyu Yang
- The Queen’s University of Belfast Joint College, China Medical University, Shenyang 110122, China;
| | - Erning Tao
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| | - Yufang Ma
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| | - Shanshan Sha
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (X.L.); (J.H.); (W.W.); (E.T.)
| |
Collapse
|
4
|
Rijal R, Gomer RH. Gallein potentiates isoniazid's ability to suppress Mycobacterium tuberculosis growth. Front Microbiol 2024; 15:1369763. [PMID: 38690363 PMCID: PMC11060752 DOI: 10.3389/fmicb.2024.1369763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb), the bacterium that causes tuberculosis (TB), can be difficult to treat because of drug tolerance. Increased intracellular polyphosphate (polyP) in Mtb enhances tolerance to antibiotics, and capsular polyP in Neisseria gonorrhoeae potentiates resistance to antimicrobials. The mechanism by which bacteria utilize polyP to adapt to antimicrobial pressure is not known. In this study, we found that Mtb adapts to the TB frontline antibiotic isoniazid (INH) by enhancing the accumulation of cellular, extracellular, and cell surface polyP. Gallein, a broad-spectrum inhibitor of the polyphosphate kinase that synthesizes polyP, prevents this INH-induced increase in extracellular and cell surface polyP levels. Gallein and INH work synergistically to attenuate Mtb's ability to grow in in vitro culture and within human macrophages. Mtb when exposed to INH, and in the presence of INH, gallein inhibits cell envelope formation in most but not all Mtb cells. Metabolomics indicated that INH or gallein have a modest impact on levels of Mtb metabolites, but when used in combination, they significantly reduce levels of metabolites involved in cell envelope synthesis and amino acid, carbohydrate, and nucleoside metabolism, revealing a synergistic effect. These data suggest that gallein represents a promising avenue to potentiate the treatment of TB.
Collapse
Affiliation(s)
- Ramesh Rijal
- Gomer Lab, Department of Biology, Texas A&M University, College Station, TX, United States
| | - Richard H. Gomer
- Gomer Lab, Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
5
|
Chugh S, Tiwari P, Suri C, Gupta SK, Singh P, Bouzeyen R, Kidwai S, Srivastava M, Rameshwaram NR, Kumar Y, Asthana S, Singh R. Polyphosphate kinase-1 regulates bacterial and host metabolic pathways involved in pathogenesis of Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2024; 121:e2309664121. [PMID: 38170746 PMCID: PMC10786269 DOI: 10.1073/pnas.2309664121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Inorganic polyphosphate (polyP) is primarily synthesized by Polyphosphate Kinase-1 (PPK-1) and regulates numerous cellular processes, including energy metabolism, stress adaptation, drug tolerance, and microbial pathogenesis. Here, we report that polyP interacts with acyl CoA carboxylases, enzymes involved in lipid biosynthesis in Mycobacterium tuberculosis. We show that deletion of ppk-1 in M. tuberculosis results in transcriptional and metabolic reprogramming. In comparison to the parental strain, the Δppk-1 mutant strain had reduced levels of virulence-associated lipids such as PDIMs and TDM. We also observed that polyP deficiency in M. tuberculosis is associated with enhanced phagosome-lysosome fusion in infected macrophages and attenuated growth in mice. Host RNA-seq analysis revealed decreased levels of transcripts encoding for proteins involved in either type I interferon signaling or formation of foamy macrophages in the lungs of Δppk-1 mutant-infected mice relative to parental strain-infected animals. Using target-based screening and molecular docking, we have identified raloxifene hydrochloride as a broad-spectrum PPK-1 inhibitor. We show that raloxifene hydrochloride significantly enhanced the activity of isoniazid, bedaquiline, and pretomanid against M. tuberculosis in macrophages. Additionally, raloxifene inhibited the growth of M. tuberculosis in mice. This is an in-depth study that provides mechanistic insights into the regulation of mycobacterial pathogenesis by polyP deficiency.
Collapse
Affiliation(s)
- Saurabh Chugh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Prabhakar Tiwari
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Charu Suri
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Sonu Kumar Gupta
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Padam Singh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Rania Bouzeyen
- Institut Pasteur de Tunis, Laboratory of Transmission, Control and Immunobiology of Infections, LRII IPT02, Tunis1002, Tunisia
| | - Saqib Kidwai
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Mitul Srivastava
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Nagender Rao Rameshwaram
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Yashwant Kumar
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad121001, India
| |
Collapse
|
6
|
Chuang YM, Alameh MG, Abouneameh S, Raduwan H, Ledizet M, Weissman D, Fikrig E. A mosquito AgTRIO mRNA vaccine contributes to immunity against malaria. NPJ Vaccines 2023; 8:88. [PMID: 37286568 PMCID: PMC10244833 DOI: 10.1038/s41541-023-00679-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Malaria begins when an infected mosquito injects saliva containing Plasmodium sporozoites into the skin of a vertebrate host. To prevent malaria, vaccination is the most effective strategy and there is an urgent need for new strategies to enhance current pathogen-based vaccines. Active or passive immunization against a mosquito saliva protein, AgTRIO, contributes to protection against Plasmodium infection of mice. In this study, we generated an AgTRIO mRNA-lipid nanoparticle (LNP) and assessed its potential usefulness as a vaccine against malaria. Immunization of mice with an AgTRIO mRNA-LNP generated a robust humoral response, including AgTRIO IgG2a isotype antibodies that have been associated with protection. AgTRIO mRNA-LNP immunized mice exposed to Plasmodium berghei-infected mosquitoes had markedly reduced initial Plasmodium hepatic infection levels and increased survival compared to control mice. In addition, as the humoral response to AgTRIO waned over 6 months, additional mosquito bites boosted the AgTRIO IgG titers, including IgG1 and IgG2a isotypes, which offers a unique advantage compared to pathogen-based vaccines. These data will aid in the generation of future malaria vaccines that may include both pathogen and vector antigens.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Mohamad-Gabriel Alameh
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selma Abouneameh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Drew Weissman
- Institute for RNA Innovation and Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
He C, Li B, Gong Z, Huang S, Liu X, Wang J, Xie J, Shi T. Polyphosphate kinase 1 is involved in formation, the morphology and ultramicrostructure of biofilm of Mycobacterium smegmatis and its survivability in macrophage. Heliyon 2023; 9:e14513. [PMID: 36967885 PMCID: PMC10034464 DOI: 10.1016/j.heliyon.2023.e14513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
The most unique characteristic of Mycobacterium tuberculosis is persistence in the human host, and the biofilm formation is related to the persistance. Polyphosphate (polyP) kinase 1 (PPK1) is conserved in Mycobacteria and is responsible for polyP synthesis. polyP is a chain molecule linked by high-energy phosphate bonds, which is considered to play a very important role in bacterial persistence. However, the relationship of PPK1 and mycobacterial biofilm formation is still adequately unclear. In current study, ppk1-deficient mutant (MT), ppk1-complemented (CT) and wild-type strains of M. smegmatis mc2 155 were used to investigate the formation, morphology and ultramicrostructure of the biofilm and to analyze the lipid levels and susceptibility to vancomycin antibiotic. And then WT, MT and CT strains were used to infect macrophages and to analyze the expression levels of various inflammatory factors, respectively. We found that PPK1 was required for M. smegmatis polyP production in vivo and polyP deficiency not only attenuated the biofilm formation, but also altered the phenotype and ultramicrostructure of the biofilm and reduced the cell lipid composition (except for C16.1 and C17.1, most of the fatty acid components from C8-C24). Moreover, the ppk1-deficient mutant was also significantly more sensitive to vancomycin which targets the cell wall, and its ability to survive in macrophages was decreased, which was related to the change of the expression level of inflammatory factors in macrophage. This study demonstrates that the PPK1 can affect the biofilm structure through affecting the content of short chain fatty acid and promote intracellular survival of M. smegmatis by altering the expression of inflammatory factors. These findings establish a basis for investigating the role of PPK1 in the persistence of M. tuberculosis, and provide clues for treating latent infection of M. tuberculosis with PPK1 as a potential drug target.
Collapse
Affiliation(s)
- Cailin He
- Medical School of Hubei Minzu University, Enshi, 445000, China
| | - Bo Li
- Medical School of Hubei Minzu University, Enshi, 445000, China
| | - Zhen Gong
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Sheng Huang
- Medical School of Hubei Minzu University, Enshi, 445000, China
- Institute of Selenium Science and Industry of Hubei Minzu University, Enshi, 445000, China
| | - Xu Liu
- Medical School of Hubei Minzu University, Enshi, 445000, China
- Institute of Selenium Science and Industry of Hubei Minzu University, Enshi, 445000, China
| | - Jiajun Wang
- Medical School of Hubei Minzu University, Enshi, 445000, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, 400715, China
- Corresponding author. Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Tingyu Shi
- Medical School of Hubei Minzu University, Enshi, 445000, China
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Enshi, 445000, China
- Institute of Selenium Science and Industry of Hubei Minzu University, Enshi, 445000, China
- Corresponding author. Medical School of Hubei Minzu University, Enshi, 445000, China.
| |
Collapse
|
8
|
Karanika S, Gordy JT, Neupane P, Karantanos T, Ruelas Castillo J, Quijada D, Comstock K, Sandhu AK, Kapoor AR, Hui Y, Ayeh SK, Tasneen R, Krug S, Danchik C, Wang T, Schill C, Markham RB, Karakousis PC. An intranasal stringent response vaccine targeting dendritic cells as a novel adjunctive therapy against tuberculosis. Front Immunol 2022; 13:972266. [PMID: 36189260 PMCID: PMC9523784 DOI: 10.3389/fimmu.2022.972266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/31/2022] [Indexed: 01/26/2023] Open
Abstract
Lengthy tuberculosis (TB) treatment is required to overcome the ability of a subpopulation of persistent Mycobacterium tuberculosis (Mtb) to remain in a non-replicating, antibiotic-tolerant state characterized by metabolic remodeling, including induction of the RelMtb-mediated stringent response. We developed a novel therapeutic DNA vaccine containing a fusion of the relMtb gene with the gene encoding the immature dendritic cell-targeting chemokine, MIP-3α/CCL20. To augment mucosal immune responses, intranasal delivery was also evaluated. We found that intramuscular delivery of the MIP-3α/relMtb (fusion) vaccine or intranasal delivery of the relMtb (non-fusion) vaccine potentiate isoniazid activity more than intramuscular delivery of the DNA vaccine expressing relMtb alone in a chronic TB mouse model (absolute reduction of Mtb burden: 0.63 log10 and 0.5 log10 colony-forming units, respectively; P=0.0002 and P=0.0052), inducing pronounced Mtb-protective immune signatures. The combined approach involving intranasal delivery of the DNA MIP-3α/relMtb fusion vaccine demonstrated the greatest mycobactericidal activity together with isoniazid when compared to each approach alone (absolute reduction of Mtb burden: 1.13 log10, when compared to the intramuscular vaccine targeting relMtb alone; P<0.0001), as well as robust systemic and local Th1 and Th17 responses. This DNA vaccination strategy may be a promising adjunctive approach combined with standard therapy to shorten curative TB treatment, and also serves as proof of concept for treating other chronic bacterial infections.
Collapse
Affiliation(s)
- Styliani Karanika
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - James T. Gordy
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pranita Neupane
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Hospital, Baltimore, MD, United States
| | - Jennie Ruelas Castillo
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Darla Quijada
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kaitlyn Comstock
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Avinaash K. Sandhu
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aakanksha R. Kapoor
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yinan Hui
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Samuel K. Ayeh
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Rokeya Tasneen
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Stefanie Krug
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Carina Danchik
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Tianyin Wang
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Courtney Schill
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard B. Markham
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
9
|
Polyphosphate Kinase Is Required for the Processes of Virulence and Persistence in Acinetobacter baumannii. Microbiol Spectr 2022; 10:e0123022. [PMID: 35867473 PMCID: PMC9430702 DOI: 10.1128/spectrum.01230-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acinetobacter baumannii, one of the most successful bacteria causing severe nosocomial infection, was identified as a top-priority pathogen by the WHO. Thus, genetic manipulations to clarify the potential targets for fighting A. baumannii resistance and virulence are vital. Polyphosphate (polyP) kinase (PPK) is conserved in nearly all bacteria and is responsible for polyP formation, which is associated with bacterial pathogenicity and antibiotic resistance. In this study, ppk1-deficient (Δppk1::Apr), ppk1-complemented (Δppk1::Apr/PJL02-ppk1), and wild-type strains of A. baumannii ATCC 17978 were used to determine the influence of PPK1 on A. baumannii virulence and persistence mainly by polyP quantification, surface motility, biofilm formation, and bacterial persistence assays. Our work found that PPK1 is indispensable for polyP formation in vivo and that the motility of the PPK1-deficient strain was significantly impaired due to the lack of a pilus-like structure typically present compared with the complemented and wild-type strains. The deficiency of PPK1 also inhibited the biofilm formation of A. baumannii and decreased bacterial persistence under stimuli of high-concentration ampicillin (Amp) treatment, H2O2 stress, heat shock, and starvation stress. Furthermore, ppk1-deficient bacterium-infected mice showed a significantly reduced bacterial load and a decreased inflammatory response. However, complementation with PPK1 effectively rescued the impaired virulence and persistence of ppk1-deficient A. baumannii. In addition, metabonomic analysis revealed that PPK1 was associated with glycerophospholipid metabolism and fatty acid biosynthesis. Taken together, our results suggest that targeting PPK1 to control A. baumannii pathogenicity and persistence is a feasible strategy to fight this pathogen. IMPORTANCEA. baumannii was identified as a top-priority pathogen by the WHO due to its antibiotic resistance. Meanwhile, the pathogenicity of A. baumannii mediated by several vital virulence factors also cannot be ignored. Here, the role of PPK1 in A. baumannii was also explored. We found that the motility ability and biofilm formation of a PPK1-deficient strain were significantly impaired. Furthermore, PPK1 was essential for its persistence maintenance to resist stimuli of high-concentration Amp treatment, H2O2 stress, heat shock, and starvation stress. Metabonomic analysis revealed that PPK1 was associated with glycerophospholipid metabolism and fatty acid biosynthesis. In addition, ppk1-deficient bacterium-infected mice showed significantly reduced bacterial loads and a decreased inflammatory responses in vivo. Together, our results suggest that PPK1 is vital for A. baumannii pathogenicity and persistence.
Collapse
|
10
|
Belardinelli JM, Verma D, Li W, Avanzi C, Wiersma CJ, Williams JT, Johnson BK, Zimmerman M, Whittel N, Angala B, Wang H, Jones V, Dartois V, de Moura VCN, Gonzalez-Juarrero M, Pearce C, Schenkel AR, Malcolm KC, Nick JA, Charman SA, Wells TNC, Podell BK, Vennerstrom JL, Ordway DJ, Abramovitch RB, Jackson M. Therapeutic efficacy of antimalarial drugs targeting DosRS signaling in Mycobacterium abscessus. Sci Transl Med 2022; 14:eabj3860. [PMID: 35196022 DOI: 10.1126/scitranslmed.abj3860] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A search for alternative Mycobacterium abscessus treatments led to our interest in the two-component regulator DosRS, which, in Mycobacterium tuberculosis, is required for the bacterium to establish a state of nonreplicating, drug-tolerant persistence in response to a variety of host stresses. We show here that the genetic disruption of dosRS impairs the adaptation of M. abscessus to hypoxia, resulting in decreased bacterial survival after oxygen depletion, reduced tolerance to a number of antibiotics in vitro and in vivo, and the inhibition of biofilm formation. We determined that three antimalarial drugs or drug candidates, artemisinin, OZ277, and OZ439, can target DosS-mediated hypoxic signaling in M. abscessus and recapitulate the phenotypic effects of genetically disrupting dosS. OZ439 displayed bactericidal activity comparable to standard-of-care antibiotics in chronically infected mice, in addition to potentiating the activity of antibiotics used in combination. The identification of antimalarial drugs as potent inhibitors and adjunct inhibitors of M. abscessus in vivo offers repurposing opportunities that could have an immediate impact in the clinic.
Collapse
Affiliation(s)
- Juan Manuel Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Deepshikha Verma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Charlotte Avanzi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Crystal J Wiersma
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John T Williams
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | | | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Nicholas Whittel
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Bhanupriya Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Han Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Victoria Jones
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - Vinicius C N de Moura
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Camron Pearce
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Alan R Schenkel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Kenneth C Malcolm
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jerry A Nick
- Department of Medicine, National Jewish Health, Denver, CO, USA.,Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | | | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | - Diane J Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Robert B Abramovitch
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
11
|
Neville N, Roberge N, Jia Z. Polyphosphate Kinase 2 (PPK2) Enzymes: Structure, Function, and Roles in Bacterial Physiology and Virulence. Int J Mol Sci 2022; 23:ijms23020670. [PMID: 35054854 PMCID: PMC8776046 DOI: 10.3390/ijms23020670] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 01/27/2023] Open
Abstract
Inorganic polyphosphate (polyP) has been implicated in an astonishing array of biological functions, ranging from phosphorus storage to molecular chaperone activity to bacterial virulence. In bacteria, polyP is synthesized by polyphosphate kinase (PPK) enzymes, which are broadly subdivided into two families: PPK1 and PPK2. While both enzyme families are capable of catalyzing polyP synthesis, PPK1s preferentially synthesize polyP from nucleoside triphosphates, and PPK2s preferentially consume polyP to phosphorylate nucleoside mono- or diphosphates. Importantly, many pathogenic bacteria such as Pseudomonas aeruginosa and Acinetobacter baumannii encode at least one of each PPK1 and PPK2, suggesting these enzymes may be attractive targets for antibacterial drugs. Although the majority of bacterial polyP studies to date have focused on PPK1s, PPK2 enzymes have also begun to emerge as important regulators of bacterial physiology and downstream virulence. In this review, we specifically examine the contributions of PPK2s to bacterial polyP homeostasis. Beginning with a survey of the structures and functions of biochemically characterized PPK2s, we summarize the roles of PPK2s in the bacterial cell, with a particular emphasis on virulence phenotypes. Furthermore, we outline recent progress on developing drugs that inhibit PPK2 enzymes and discuss this strategy as a novel means of combatting bacterial infections.
Collapse
|
12
|
Gupta KR, Arora G, Mattoo A, Sajid A. Stringent Response in Mycobacteria: From Biology to Therapeutic Potential. Pathogens 2021; 10:pathogens10111417. [PMID: 34832573 PMCID: PMC8622095 DOI: 10.3390/pathogens10111417] [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: 08/31/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis is a human pathogen that can thrive inside the host immune cells for several years and cause tuberculosis. This is due to the propensity of M. tuberculosis to synthesize a sturdy cell wall, shift metabolism and growth, secrete virulence factors to manipulate host immunity, and exhibit stringent response. These attributes help M. tuberculosis to manage the host response, and successfully establish and maintain an infection even under nutrient-deprived stress conditions for years. In this review, we will discuss the importance of mycobacterial stringent response under different stress conditions. The stringent response is mediated through small signaling molecules called alarmones “(pp)pGpp”. The synthesis and degradation of these alarmones in mycobacteria are mediated by Rel protein, which is both (p)ppGpp synthetase and hydrolase. Rel is important for all central dogma processes—DNA replication, transcription, and translation—in addition to regulating virulence, drug resistance, and biofilm formation. Rel also plays an important role in the latent infection of M. tuberculosis. Here, we have discussed the literature on alarmones and Rel proteins in mycobacteria and highlight that (p)ppGpp-analogs and Rel inhibitors could be designed and used as antimycobacterial compounds against M. tuberculosis and non-tuberculous mycobacterial infections.
Collapse
Affiliation(s)
| | - Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Abid Mattoo
- Pharmaceutical Development, Ultragenyx Gene Therapy, Woburn, MA 01801, USA;
| | - Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
- Correspondence: or
| |
Collapse
|
13
|
Danchik C, Wang S, Karakousis PC. Targeting the Mycobacterium tuberculosis Stringent Response as a Strategy for Shortening Tuberculosis Treatment. Front Microbiol 2021; 12:744167. [PMID: 34690990 PMCID: PMC8529327 DOI: 10.3389/fmicb.2021.744167] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
The stringent response is well conserved across bacterial species and is a key pathway involved both in bacterial survival and virulence and in the induction of antibiotic tolerance in Mycobacteria. It is mediated by the alarmone (p)ppGpp and the regulatory molecule inorganic polyphosphate in response to stress conditions such as nutrient starvation. Efforts to pharmacologically target various components of the stringent response have shown promise in modulating mycobacterial virulence and antibiotic tolerance. In this review, we summarize the current understanding of the stringent response and its role in virulence and tolerance in Mycobacteria, including evidence that targeting this pathway could have therapeutic benefit.
Collapse
Affiliation(s)
| | | | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
14
|
Albumin fusion with granulocyte-macrophage colony-stimulating factor acts as an immunotherapy against chronic tuberculosis. Cell Mol Immunol 2021; 18:2393-2401. [PMID: 32382128 PMCID: PMC8484439 DOI: 10.1038/s41423-020-0439-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
A long duration of treatment and emerging drug resistance pose significant challenges for global tuberculosis (TB) eradication efforts. Therefore, there is an urgent need to develop novel strategies to shorten TB treatment regimens and to treat drug-resistant TB. Using an albumin-fusion strategy, we created a novel albumin-fused granulocyte-macrophage colony-stimulating factor (albGM-CSF) molecule that harnesses albumin's long half-life and targeting abilities to enhance the biostability of GM-CSF and direct it to the lymph nodes, where the effects of GM-CSF can increase dendritic cell populations crucial for eliciting a potent immune response. In this study, we demonstrate that albGM-CSF serves as a novel immunotherapy for chronic Mycobacterium tuberculosis (Mtb) infections by enhancing GM-CSF biostability in serum. Specifically, albumin is very safe, stable, and has a long half-life, thereby enhancing the biostability of GM-CSF. In the lungs and draining lymph nodes, albGM-CSF is able to increase the numbers of dendritic cells, which are crucial for the activation of naive T cells and for eliciting potent immune responses. Subcutaneous administration of albGM-CSF alone reduced the mean lung bacillary burden in mice with chronic tuberculosis infection. While GM-CSF administration was associated with IL-1β release from Mtb-infected dendritic cells and macrophages, higher IL-1β levels were observed in albGM-CSF-treated mice with chronic tuberculosis infection than in mice receiving GM-CSF. Albumin fusion with GM-CSF represents a promising strategy for the control of chronic lung tuberculosis infections and serves as a novel therapeutic vaccination platform for other infectious diseases and malignancies.
Collapse
|
15
|
Park HE, Lee W, Shin MK, Shin SJ. Understanding the Reciprocal Interplay Between Antibiotics and Host Immune System: How Can We Improve the Anti-Mycobacterial Activity of Current Drugs to Better Control Tuberculosis? Front Immunol 2021; 12:703060. [PMID: 34262571 PMCID: PMC8273550 DOI: 10.3389/fimmu.2021.703060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, remains a global health threat despite recent advances and insights into host-pathogen interactions and the identification of diverse pathways that may be novel therapeutic targets for TB treatment. In addition, the emergence and spread of multidrug-resistant Mtb strains led to a low success rate of TB treatments. Thus, novel strategies involving the host immune system that boost the effectiveness of existing antibiotics have been recently suggested to better control TB. However, the lack of comprehensive understanding of the immunomodulatory effects of anti-TB drugs, including first-line drugs and newly introduced antibiotics, on bystander and effector immune cells curtailed the development of effective therapeutic strategies to combat Mtb infection. In this review, we focus on the influence of host immune-mediated stresses, such as lysosomal activation, metabolic changes, oxidative stress, mitochondrial damage, and immune mediators, on the activities of anti-TB drugs. In addition, we discuss how anti-TB drugs facilitate the generation of Mtb populations that are resistant to host immune response or disrupt host immunity. Thus, further understanding the interplay between anti-TB drugs and host immune responses may enhance effective host antimicrobial activities and prevent Mtb tolerance to antibiotic and immune attacks. Finally, this review highlights novel adjunctive therapeutic approaches against Mtb infection for better disease outcomes, shorter treatment duration, and improved treatment efficacy based on reciprocal interactions between current TB antibiotics and host immune cells.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
16
|
Fernández L, Gutiérrez D, García P, Rodríguez A. Environmental pH is a key modulator of Staphylococcus aureus biofilm development under predation by the virulent phage phiIPLA-RODI. ISME JOURNAL 2020; 15:245-259. [PMID: 32963343 DOI: 10.1038/s41396-020-00778-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/03/2020] [Accepted: 09/14/2020] [Indexed: 11/09/2022]
Abstract
Previous work had shown that, in some Staphylococcus aureus strains, low concentrations of the virulent phage vB_SauM_phiIPLA-RODI (phiIPLA-RODI) promoted the formation of DNA-rich biofilms, whose cells exhibited significant transcriptional differences compared to an uninfected control. This study aimed to dissect the sequence of events leading to these changes. Analysis of phage propagation throughout biofilm development revealed that the number of phage particles increased steadily up to a certain point and then declined. This partial phage inactivation seemed to be a consequence of medium acidification due to glucose fermentation by the bacterium. Computer simulation of phage-host dynamics during biofilm development showed how even small differences in pH evolution can affect the outcome of phage infection. An acidic pH, together with successful phage propagation, was also necessary to observe the phage-associated changes in biofilm architecture and in the transcriptional profile of the bacterial population. Altogether, this study shows how the dynamics between phage and host can be tightly coordinated through an environmental cue, even in the context of a complex biofilm population.
Collapse
Affiliation(s)
- Lucía Fernández
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain. .,DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.
| | - Diana Gutiérrez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.,Laboratory of Applied Biotechnology, Department of Applied Biosciences, Faculty of Bioscience engineering, Ghent University, 9000, Ghent, Belgium
| | - Pilar García
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.,DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ana Rodríguez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Paseo Río Linares s/n, 33300, Villaviciosa, Asturias, Spain.,DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
17
|
Boldrin F, Provvedi R, Cioetto Mazzabò L, Segafreddo G, Manganelli R. Tolerance and Persistence to Drugs: A Main Challenge in the Fight Against Mycobacterium tuberculosis. Front Microbiol 2020; 11:1924. [PMID: 32983003 PMCID: PMC7479264 DOI: 10.3389/fmicb.2020.01924] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
The treatment of tuberculosis is extremely long. One of the reasons why Mycobacterium tuberculosis elimination from the organism takes so long is that in particular environmental conditions it can become tolerant to drugs and/or develop persisters able to survive killing even from very high drug concentrations. Tolerance develops in response to a harsh environment exposure encountered by bacteria during infection, mainly due to the action of the immune system, whereas persistence results from the presence of heterogeneous bacterial populations with different degrees of drug sensitivity, and can be induced by exposure to stress conditions. Here, we review the actual knowledge on the stress response mechanisms enacted by M. tuberculosis during infection, which leads to increased drug tolerance or development of a highly drug-resistant subpopulation.
Collapse
Affiliation(s)
- Francesca Boldrin
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | - Greta Segafreddo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | |
Collapse
|
18
|
Chuang YM, Dutta NK, Gordy JT, Campodónico VL, Pinn ML, Markham RB, Hung CF, Karakousis PC. Antibiotic Treatment Shapes the Antigenic Environment During Chronic TB Infection, Offering Novel Targets for Therapeutic Vaccination. Front Immunol 2020; 11:680. [PMID: 32411131 PMCID: PMC7198710 DOI: 10.3389/fimmu.2020.00680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/26/2020] [Indexed: 12/03/2022] Open
Abstract
The lengthy and complicated current regimen required to treat drug-susceptible tuberculosis (TB) reflects the ability of Mycobacterium tuberculosis (Mtb) to persist in host tissues. The stringent response pathway, governed by the dual (p)ppGpp synthetase/hydrolase, RelMtb, is a major mechanism underlying Mtb persistence and antibiotic tolerance. In the current study, we addressed the hypothesis that RelMtb is a “persistence antigen” presented during TB chemotherapy and that enhanced immunity to RelMtb can enhance the tuberculocidal activity of the first-line anti-TB drug, isoniazid, which has reduced efficacy against Mtb persisters. C57BL/6 mice and Hartley guinea pigs were aerosol-infected with M. tuberculosis (Mtb) and, 4 weeks later, received either human-equivalent daily doses of isoniazid alone, or isoniazid in combination with a DNA vaccine targeting relMtb. After isoniazid treatment, there was a significant reduction in dominant antigen ESAT6-reactive CD4+ or TB10.4-reactive CD8+ T cells in the lungs and spleens of mice. However, the total number of RelMtb-reactive CD4+ T cells remained stable in mouse lungs and spleens, as did the number of RelMtb-reactive CD8+T cells. Therapeutic vaccination with relMtb DNA vaccine enhanced the activity of isoniazid in Mtb-infected C57BL/6 mice and guinea pigs. When treatment with isoniazid was discontinued, mice immunized with the relMtb DNA vaccine showed a lower mean lung bacterial burden at relapse compared to the control group. Our work shows that antitubercular treatment shapes the antigenic environment, and that therapeutic vaccination targeting the Mtb stringent response may represent a novel approach to enhance immunity against Mtb persisters, with the ultimate goal of shortening curative TB treatment.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Noton K Dutta
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - James T Gordy
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Victoria L Campodónico
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael L Pinn
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard B Markham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
19
|
Andreeva N, Ledova L, Ryazanova L, Tomashevsky A, Kulakovskaya T, Eldarov M. Ppn2 endopolyphosphatase overexpressed in Saccharomyces cerevisiae: Comparison with Ppn1, Ppx1, and Ddp1 polyphosphatases. Biochimie 2019; 163:101-107. [DOI: 10.1016/j.biochi.2019.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022]
|
20
|
Shahbaaz M, Nkaule A, Christoffels A. Designing novel possible kinase inhibitor derivatives as therapeutics against Mycobacterium tuberculosis: An in silico study. Sci Rep 2019; 9:4405. [PMID: 30867456 PMCID: PMC6416319 DOI: 10.1038/s41598-019-40621-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/18/2019] [Indexed: 11/30/2022] Open
Abstract
Rv2984 is one of the polyphosphate kinases present in Mycobacterium tuberculosis involved in the catalytic synthesis of inorganic polyphosphate, which plays an essential role in bacterial virulence and drug resistance. Consequently, the structure of Rv2984 was investigated and an 18 membered compound library was designed by altering the scaffolds of computationally identified inhibitors. The virtual screening of these altered inhibitors was performed against Rv2984 and the top three scoring inhibitors were selected, exhibiting the free energy of binding between 8.2–9 kcal mol−1 and inhibition constants in the range of 255–866 nM. These selected molecules showed relatively higher binding affinities against Rv2984 compared to the first line drugs Isoniazid and Rifampicin. Furthermore, the docked complexes were further analyzed in explicit water conditions using 100 ns Molecular Dynamics simulations. Through the assessment of obtained trajectories, the interactions between the protein and selected inhibitors including first line drugs were evaluated using MM/PBSA technique. The results validated the higher efficiency of the designed molecules compared to 1st line drugs with total interaction energies observed between −100 kJ mol−1 and −1000 kJ mol−1. This study will facilitate the process of drug designing against M. tuberculosis and can be used in the development of potential therapeutics against drug-resistant strains of bacteria.
Collapse
Affiliation(s)
- Mohd Shahbaaz
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Anati Nkaule
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa
| | - Alan Christoffels
- South African National Bioinformatics Institute (SANBI), SA Medical Research Council Bioinformatics Unit, University of the Western Cape, Private Bag X17, Bellville, 7535, Cape Town, South Africa.
| |
Collapse
|
21
|
Srisanga K, Suthapot P, Permsirivisarn P, Govitrapong P, Tungpradabkul S, Wongtrakoongate P. Polyphosphate kinase 1 of Burkholderia pseudomallei controls quorum sensing, RpoS and host cell invasion. J Proteomics 2019; 194:14-24. [DOI: 10.1016/j.jprot.2018.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/16/2018] [Accepted: 12/25/2018] [Indexed: 12/18/2022]
|
22
|
Gautam LK, Sharma P, Capalash N. Bacterial Polyphosphate Kinases Revisited: Role in Pathogenesis and Therapeutic Potential. Curr Drug Targets 2019; 20:292-301. [DOI: 10.2174/1389450119666180801120231] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/02/2018] [Accepted: 07/31/2018] [Indexed: 11/22/2022]
Abstract
Bacterial infections have always been an unrestrained challenge to the medical community due to the rise of multi-drug tolerant and resistant strains. Pioneering work on Escherichia coli polyphosphate kinase (PPK) by Arthur Kornberg has generated great interest in this polyphosphate (PolyP) synthesizing enzyme. PPK has wide distribution among pathogens and is involved in promoting pathogenesis, stress management and susceptibility to antibiotics. Further, the absence of a PPK orthologue in humans makes it a potential drug target. This review covers the functional and structural aspects of polyphosphate kinases in bacterial pathogens. A description of molecules being designed against PPKs has been provided, challenges associated with PPK inhibitor design are highlighted and the strategies to enable development of efficient drug against this enzyme have also been discussed.
Collapse
Affiliation(s)
- Lalit Kumar Gautam
- Department of Biotechnology, Panjab University, BMS Block-I, Sector- 25, Chandigarh, 160014, India
| | - Prince Sharma
- Department of Microbiology, Panjab University, BMS Block-I, Sector- 25, Chandigarh, 160014, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, BMS Block-I, Sector- 25, Chandigarh, 160014, India
| |
Collapse
|
23
|
Nakamura A, Kawano N, Motomura K, Kuroda A, Sekiguchi K, Miyado M, Kang W, Miyamoto Y, Hanai M, Iwai M, Yamada M, Hamatani T, Saito T, Saito H, Tanaka M, Umezawa A, Miyado K. Degradation of phosphate polymer polyP enhances lactic fermentation in mice. Genes Cells 2018; 23:904-914. [PMID: 30144248 DOI: 10.1111/gtc.12639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 01/10/2023]
Abstract
In bacteria, a polymer of inorganic phosphate (Pi) (inorganic polyphosphate; polyP) is enzymatically produced and consumed as an alternative phosphate donor for adenosine triphosphate (ATP) production to protect against nutrient starvation. In vertebrates, polyP has been dismissed as a "molecular fossil" due to the lack of any known physiological function. Here, we have explored its possible role by producing transgenic (TG) mice widely expressing Saccharomyces cerevisiae exopolyphosphatase 1 (ScPPX1), which catalyzes hydrolytic polyP degradation. TG mice were produced and displayed reduced mitochondrial respiration in muscles. In female TG mice, the blood concentration of lactic acid was enhanced, whereas ATP storage in liver and brain tissues was reduced significantly. Thus, we suggested that the elongation of polyP reduces the intracellular Pi concentration, suppresses anaerobic lactic acid production, and sustains mitochondrial respiration. Our results provide an insight into the physiological role of polyP in mammals, particularly in females.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku, Tokyo, Japan.,Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Natsuko Kawano
- Department of Life Sciences, School of Agriculture, Meiji University, Tama, Kawasaki, Kanagawa, Japan
| | - Kei Motomura
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Akio Kuroda
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | | | - Mami Miyado
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Yoshitaka Miyamoto
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Maito Hanai
- Department of Life Sciences, School of Agriculture, Meiji University, Tama, Kawasaki, Kanagawa, Japan
| | - Maki Iwai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Mitsutoshi Yamada
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Toshio Hamatani
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Takakazu Saito
- Department of Perinatal Medicine and Maternal Care, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Hidekazu Saito
- Department of Perinatal Medicine and Maternal Care, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| |
Collapse
|
24
|
Bhaskar A, De Piano C, Gelman E, McKinney JD, Dhar N. Elucidating the role of (p)ppGpp in mycobacterial persistence against antibiotics. IUBMB Life 2018; 70:836-844. [PMID: 30092117 DOI: 10.1002/iub.1888] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/23/2018] [Indexed: 01/05/2023]
Abstract
Bacterial persistence, the ability of bacteria to survive high concentrations of antibiotics for extended periods of time, is an important contributing factor to therapy failure and development of chronic and recurrent infections. Several recent studies have suggested that this persistence is mediated primarily by (p)ppGpp, through its interactions with toxin-antitoxin modules and polyphosphates. In this study, we address whether these key players play a role in mycobacterial persistence against antibiotics. We targeted these specific pathways in Mycobacterium smegmatis by constructing deletion strains of (p)ppGpp synthetase/hydrolase (relA), polyphosphate kinases (ppk1 and ppk2), exopolyphosphatases (ppx1 and ppx2), and the lon protease. None of these mutant strains exhibited altered levels of persisters against isoniazid and ciprofloxacin, when compared with wild-type strain. Even under conditions in which the stringent response usually gets activated, these strains displayed wild-type persister levels. Interestingly, we also found that unlike Escherichia coli, maintaining M. smegmatis in exponential phase by repeated passaging does not eliminate persisters suggesting that at least against the antibiotics tested, stationary-phase dependent persisters (type I) are not the major contributors. Thus, our data demonstrate that multiple mechanisms of antibiotic persistence exist and that these vary widely among different bacterial species. © 2018 IUBMB Life, 70(9):836-844, 2018.
Collapse
Affiliation(s)
- Ashima Bhaskar
- Laboratory of Microbiology and Microtechnology, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Cyntia De Piano
- Laboratory of Microbiology and Microtechnology, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Ekaterina Gelman
- Laboratory of Microbiology and Microtechnology, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - John D McKinney
- Laboratory of Microbiology and Microtechnology, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Neeraj Dhar
- Laboratory of Microbiology and Microtechnology, School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
25
|
Yang Y, Richards JP, Gundrum J, Ojha AK. GlnR Activation Induces Peroxide Resistance in Mycobacterial Biofilms. Front Microbiol 2018; 9:1428. [PMID: 30022971 PMCID: PMC6039565 DOI: 10.3389/fmicb.2018.01428] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/11/2018] [Indexed: 12/31/2022] Open
Abstract
Mycobacteria spontaneously form surface-associated multicellular communities, called biofilms, which display resistance to a wide range of exogenous stresses. A causal relationship between biofilm formation and emergence of stress resistance is not known. Here, we report that activation of a nitrogen starvation response regulator, GlnR, during the development of Mycobacterium smegmatis biofilms leads to peroxide resistance. The resistance arises from induction of a GlnR-dependent peroxide resistance (gpr) gene cluster comprising of 8 ORFs (MSMEG_0565-0572). Expression of gpr increases the NADPH to NADP ratio, suggesting that a reduced cytosolic environment of nitrogen-starved cells in biofilms contributes to peroxide resistance. Increased NADPH levels from gpr activity likely support the activity of enzymes involved in nitrogen assimilation, as suggested by a higher threshold of nitrogen supplement required by a gpr mutant to form biofilms. Together, our study uniquely interlinks a nutrient sensing mechanism with emergence of stress resistance during mycobacterial biofilm development. The gpr gene cluster is conserved in several mycobacteria that can cause nosocomial infections, offering a possible explanation for their resistance to peroxide-based sterilization of medical equipment.
Collapse
Affiliation(s)
- Yong Yang
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Jacob P. Richards
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer Gundrum
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, United States
| | - Anil K. Ojha
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, United States
- Department of Biomedical Sciences, University at Albany, Albany, NY, United States
| |
Collapse
|
26
|
Prusa J, Zhu DX, Stallings CL. The stringent response and Mycobacterium tuberculosis pathogenesis. Pathog Dis 2018; 76:5035815. [PMID: 29947752 PMCID: PMC7191866 DOI: 10.1093/femspd/fty054] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022] Open
Abstract
During infection, the host restrains Mycobacterium tuberculosis (Mtb) from proliferating by imposing an arsenal of stresses. Despite this onslaught of attacks, Mtb is able to persist for the lifetime of the host, indicating that this pathogen has substantial molecular mechanisms to resist host-inflicted damage. The stringent response is a conserved global stress response in bacteria that involves the production of the hyperphosphorylated guanine nucleotides ppGpp and pppGpp (collectively called (p)ppGpp). (p)ppGpp then regulates a number of cellular processes to adjust the physiology of the bacteria to promote survival in different environments. Survival in the presence of host-generated stresses is an essential quality of successful pathogens, and the stringent response is critical for the intracellular survival of a number of pathogenic bacteria. In addition, the stringent response has been linked to virulence gene expression, persistence, latency and drug tolerance. In Mtb, (p)ppGpp synthesis is required for survival in low nutrient conditions, long term culture and during chronic infection in animal models, all indicative of a strict requirement for (p)ppGpp during exposure to stresses associated with infection. In this review we discuss (p)ppGpp metabolism and how this functions as a critical regulator of Mtb virulence.
Collapse
Affiliation(s)
- Jerome Prusa
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Dennis X Zhu
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
27
|
Antonova AV, Gryadunov DA, Zimenkov DV. Molecular Mechanisms of Drug Tolerance in Mycobacterium tuberculosis. Mol Biol 2018. [DOI: 10.1134/s0026893318030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Dow A, Prisic S. Alternative ribosomal proteins are required for growth and morphogenesis of Mycobacterium smegmatis under zinc limiting conditions. PLoS One 2018; 13:e0196300. [PMID: 29684089 PMCID: PMC5912738 DOI: 10.1371/journal.pone.0196300] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/10/2018] [Indexed: 01/19/2023] Open
Abstract
Zinc is an essential micronutrient required for proper structure and function of many proteins. Bacteria regularly encounter zinc depletion and have evolved diverse mechanisms to continue growth when zinc is limited, including the expression of zinc-independent paralogs of zinc-binding proteins. Mycobacteria have a conserved operon encoding four zinc-independent alternative ribosomal proteins (AltRPs) that are expressed when zinc is depleted. It is unknown if mycobacterial AltRPs replace their primary paralogs in the ribosome and maintain protein synthesis under zinc-limited conditions, and if such replacements contribute to their physiology. This study shows that AltRPs from Mycobacterium smegmatis are essential for growth when zinc ion is scarce. Specifically, the deletion mutant of this operon (ΔaltRP) is unable to grow in media containing a high-affinity zinc chelator, while growth of the wild type strain is unaffected under the same conditions. However, when zinc is gradually depleted during growth in zinc-limited medium, the ΔaltRP mutant maintains the same growth rate as seen for the wild type strain. In contrast to M. smegmatis grown with sufficient zinc supplementation that forms shorter cells when transitioning from logarithmic to stationary phase, M. smegmatis deficient for zinc elongates after the expression of AltRPs in late logarithmic phase. These zinc-depleted bacteria also exhibit a remarkable morphology characterized by a condensed chromosome, increased number of polyphosphate granules, and distinct appearance of lipid bodies and the cell wall compared to the zinc-replete cells. However, the ΔaltRP cells fail to elongate and transition into the zinc-limited morphotype, resembling the wild type zinc-replete bacteria instead. Therefore, the altRP operon in M. smegmatis has a vital role in continuation of growth when zinc is scarce and in triggering specific morphogenesis during the adaptation to zinc limitation, suggesting that AltRPs can functionally replace their zinc-dependent paralogs, but also contribute to mycobacterial physiology in a unique way.
Collapse
Affiliation(s)
- Allexa Dow
- Department of Microbiology, University of Hawai‛i at Mānoa, Honolulu, Hawai‛i, United States of America
| | - Sladjana Prisic
- Department of Microbiology, University of Hawai‛i at Mānoa, Honolulu, Hawai‛i, United States of America
- * E-mail:
| |
Collapse
|
29
|
Wang Z, Cumming BM, Mao C, Zhu Y, Lu P, Steyn AJC, Chen S, Hu Y. RbpA and σ B association regulates polyphosphate levels to modulate mycobacterial isoniazid-tolerance. Mol Microbiol 2018; 108:627-640. [PMID: 29575247 DOI: 10.1111/mmi.13952] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2018] [Indexed: 12/13/2022]
Abstract
To facilitate survival under drug stresses, a small population of Mycobacterium tuberculosis can tolerate bactericidal concentrations of drugs without genetic mutations. These drug-tolerant mycobacteria can be induced by environmental stresses and contribute to recalcitrant infections. However, mechanisms underlying the development of drug-tolerant mycobacteria remain obscure. Herein, we characterized a regulatory pathway which is important for the tolerance to isoniazid (INH) in Mycobacterium smegmatis. We found that the RNA polymerase binding protein RbpA associates with the stress response sigma factor σB , to activate the transcription of ppk1, the gene encoding polyphosphate kinase. Subsequently, intracellular levels of inorganic polyphosphate increase to promote INH-tolerant mycobacteria. Interestingly, σB and ppk1 expression varied proportionately in mycobacterial populations and positively correlated with tolerance to INH in individual mycobacteria. Moreover, sigB and ppk1 transcription are both induced upon nutrient depletion, a condition that stimulates the formation of INH-tolerant mycobacteria. Over-expression of ppk1 in rbpA knockdown or sigB deleted strains successfully restored the number of INH-tolerant mycobacteria under both normal growth and nutrient starved conditions. These data suggest that RbpA and σB regulate ppk1 expression to control drug tolerance both during the logarithmic growth phase and under the nutrition starved conditions.
Collapse
Affiliation(s)
- Zhongwei Wang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | | | - Chunyou Mao
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan Zhu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Pei Lu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Adrie J C Steyn
- Africa Health Research Institute, Durban, South Africa.,Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Shiyun Chen
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yangbo Hu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
30
|
Zondervan NA, van Dam JCJ, Schaap PJ, Martins Dos Santos VAP, Suarez-Diez M. Regulation of Three Virulence Strategies of Mycobacterium tuberculosis: A Success Story. Int J Mol Sci 2018; 19:E347. [PMID: 29364195 PMCID: PMC5855569 DOI: 10.3390/ijms19020347] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 01/19/2018] [Accepted: 01/21/2018] [Indexed: 12/28/2022] Open
Abstract
Tuberculosis remains one of the deadliest diseases. Emergence of drug-resistant and multidrug-resistant M. tuberculosis strains makes treating tuberculosis increasingly challenging. In order to develop novel intervention strategies, detailed understanding of the molecular mechanisms behind the success of this pathogen is required. Here, we review recent literature to provide a systems level overview of the molecular and cellular components involved in divalent metal homeostasis and their role in regulating the three main virulence strategies of M. tuberculosis: immune modulation, dormancy and phagosomal rupture. We provide a visual and modular overview of these components and their regulation. Our analysis identified a single regulatory cascade for these three virulence strategies that respond to limited availability of divalent metals in the phagosome.
Collapse
Affiliation(s)
- Niels A Zondervan
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| | - Jesse C J van Dam
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| | - Peter J Schaap
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| | - Vitor A P Martins Dos Santos
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
- LifeGlimmer GmbH, Markelstrasse 38, 12163 Berlin, Germany.
| | - Maria Suarez-Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, Stippeneng 4, 6708 WE Wageningen, The Netherlands.
| |
Collapse
|
31
|
New Approaches and Therapeutic Options for Mycobacterium tuberculosis in a Dormant State. Clin Microbiol Rev 2017; 31:31/1/e00060-17. [PMID: 29187395 DOI: 10.1128/cmr.00060-17] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We are far away from the days when tuberculosis (TB) accounted for 1 in 4 deaths during the 19th century. However, Mycobacterium tuberculosis complex (MTBC) strains are still the leading cause of morbidity and mortality by a single infectious disease, with 9.6 million cases and 1.5 million deaths reported. One-third of the world's population is estimated by the WHO to be infected with latent TB. During the last decade, several studies have aimed to define the characteristics of dormant bacteria in these latent infections. General features of the shift to a dormant state encompass several phenotypic changes that reduce metabolic activity. This low metabolic state is thought to increase the resistance of MTBC strains to host/environmental stresses, including antibiotic action. Once the stress ceases (e.g., interruption of treatment), dormant cells can reactivate and cause symptomatic disease again. Therefore, a proper understanding of dormancy could guide the rational development of new treatment regimens that target dormant cells, reducing later relapse. Here, we briefly summarize the latest data on the genetics involved in the regulation of dormancy and discuss new approaches to TB treatment.
Collapse
|
32
|
Cabello FC, Godfrey HP, Bugrysheva J, Newman SA. Sleeper cells: the stringent response and persistence in the Borreliella (Borrelia) burgdorferi enzootic cycle. Environ Microbiol 2017; 19:3846-3862. [PMID: 28836724 PMCID: PMC5794220 DOI: 10.1111/1462-2920.13897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/12/2017] [Accepted: 08/17/2017] [Indexed: 12/27/2022]
Abstract
Infections with tick-transmitted Borreliella (Borrelia) burgdorferi, the cause of Lyme disease, represent an increasingly large public health problem in North America and Europe. The ability of these spirochetes to maintain themselves for extended periods of time in their tick vectors and vertebrate reservoirs is crucial for continuance of the enzootic cycle as well as for the increasing exposure of humans to them. The stringent response mediated by the alarmone (p)ppGpp has been determined to be a master regulator in B. burgdorferi. It modulates the expression of identified and unidentified open reading frames needed to deal with and overcome the many nutritional stresses and other challenges faced by the spirochete in ticks and animal reservoirs. The metabolic and morphologic changes resulting from activation of the stringent response in B. burgdorferi may also be involved in the recently described non-genetic phenotypic phenomenon of tolerance to otherwise lethal doses of antimicrobials and to other antimicrobial activities. It may thus constitute a linchpin in multiple aspects of infections with Lyme disease borrelia, providing a link between the micro-ecological challenges of its enzootic life-cycle and long-term residence in the tissues of its animal reservoirs, with the evolutionary side effect of potential persistence in incidental human hosts.
Collapse
Affiliation(s)
- Felipe C. Cabello
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
| | - Henry P. Godfrey
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Julia Bugrysheva
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stuart A. Newman
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
33
|
彭 亮, 区 静, 潘 嘉, 邓 聪, 陈 景, 曹 虹. [Expression of Proteus mirabilis polyphosphate kinase and preparation of its polyclonal antibodies]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:312-316. [PMID: 28377345 PMCID: PMC6780429 DOI: 10.3969/j.issn.1673-4254.2017.03.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To express and purify polyphosphate kinase (PPK) from Proteus mirabilis and prepare the polyclonal antibody against PPK. METHODS The antigenicity and hydrophobicity of PPK were analyzed using software. The N-terminal conservative sequence containing 309 amino acids was selected as the target peptide, and its corresponding gene sequence with modification based on prokaryotic cells-preferred codon was synthesized and inserted into plasmid pET28b(+). The constructed recombinant plasmid was transformed into Escherichia coli BL21 (DE3) and induced with IPTG. The expressed fusion protein was purified using Ni-affinity chromatography. The purified protein was injected along with adjuvant in rabbits to prepare the polyclonal antibodies against PPK. RESULTS AND CONCLUSION PPK fusion protein expressed by E. coli was purified successfully using Ni-affinity chromatography. ELISA result demonstrated that the harvested rabbit anti-sera against PPK had a high titer of 1:512 000, and Western blotting showed a good specificity of the antibody, which can be used further study of the role of PPK in the pathogenesis of Proteus mirabilis infection.
Collapse
Affiliation(s)
- 亮 彭
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 静怡 区
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 嘉韵 潘
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 聪 邓
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 景红 陈
- 广州医科大学附属第二医院 血液科,广东 广州 510260Department of Hematology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 虹 曹
- 南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research/ School of Public Health, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
34
|
彭 亮, 区 静, 潘 嘉, 邓 聪, 陈 景, 曹 虹. [Expression of Proteus mirabilis polyphosphate kinase and preparation of its polyclonal antibodies]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:312-316. [PMID: 28377345 PMCID: PMC6780429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To express and purify polyphosphate kinase (PPK) from Proteus mirabilis and prepare the polyclonal antibody against PPK. METHODS The antigenicity and hydrophobicity of PPK were analyzed using software. The N-terminal conservative sequence containing 309 amino acids was selected as the target peptide, and its corresponding gene sequence with modification based on prokaryotic cells-preferred codon was synthesized and inserted into plasmid pET28b(+). The constructed recombinant plasmid was transformed into Escherichia coli BL21 (DE3) and induced with IPTG. The expressed fusion protein was purified using Ni-affinity chromatography. The purified protein was injected along with adjuvant in rabbits to prepare the polyclonal antibodies against PPK. RESULTS AND CONCLUSION PPK fusion protein expressed by E. coli was purified successfully using Ni-affinity chromatography. ELISA result demonstrated that the harvested rabbit anti-sera against PPK had a high titer of 1:512 000, and Western blotting showed a good specificity of the antibody, which can be used further study of the role of PPK in the pathogenesis of Proteus mirabilis infection.
Collapse
Affiliation(s)
- 亮 彭
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 静怡 区
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 嘉韵 潘
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 聪 邓
- 广州医科大学附属第二医院 检验科,广东 广州 510260Department of Clinical Laboratory, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 景红 陈
- 广州医科大学附属第二医院 血液科,广东 广州 510260Department of Hematology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - 虹 曹
- 南方医科大学公共卫生学院//广东省热带病研究重点实验室,微生物学系,广东 广州 510515Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research/ School of Public Health, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|