101
|
Liu DQ, Zhang JL, Pan ZF, Mai JT, Mei HJ, Dai Y, Zhang L, Wang QZ. Over-expression of Tgs1 in Mycobacterium marinum enhances virulence in adult zebrafish. Int J Med Microbiol 2019; 310:151378. [PMID: 31757695 DOI: 10.1016/j.ijmm.2019.151378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/28/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), can persist in the host for decades without causing TB symptoms and can cause a latent infection, which is an intricate challenge of current TB control. The DosR regulon, which contains approximately 50 genes, is crucial in the non-replicating persistence of Mtb. tgs1 is one of the most powerfully induced genes in this regulon during Mtb non-replicating persistence. The gene encodes a triacyl glycerol synthase catalyzing synthesis of triacyl glycerol (TAG), which is proposed as an energy source during bacilli persistence. Here, western blotting showed that the Tgs1 protein was upregulated in clinical Mtb strains. To detect its physiological effects on mycobacterium, we constructed serial recombinant M. marinum including over-expressed Tgs1(Tgs1-H), reduced-expressed Tgs1(Tgs1-L), and wild type M. marinum strains as controls. Tgs1 over-expression did not influence M. marinum growth under aerobic shaking and in hypoxic cultures, while growth advantages were observed at an early stage under nutrient starvation. Transmission electron microscopy revealed more lipid droplets in Tgs1-H than the other two strains; the droplets filled the cytoplasm. Two-dimensional thin-layer chromatography revealed more phosphatidyl-myo-inositol mannosides in the Tgs1-H cell wall. To assess the virulence of recombinant M. marinum in the natural host, adult zebrafish were infected with Tgs1-H or wild type strains. Hypervirulence of Tgs1-H was characterized by markedly increased bacterial load and early death of adult zebrafish. Remarkably, zebrafish infected with Tgs1-H developed necrotizing granulomas much more rapidly and in higher amounts, which facilitated mycobacterial replication and dissemination among organs and eventual tissue destruction in zebrafish. RNA sequencing analysis showed Tgs1-H induced 13 genes differentially expressed under aerobiosis. Among them, PE_PGRS54 (MMAR_5307),one of the PE_PGRS family of antigens, was markedly up-regulated, while 110 coding genes were down-regulated in Tgs1-L.The 110 genes included 22 member genes of the DosR regulon. The collective results indicate an important role for the Tgs1 protein of M. marinumin progression of infection in the natural host. Tgs1 signaling may be involved in a previously unknown behavior of M. marinum under hypoxia/aerobiosis.
Collapse
Affiliation(s)
- Ding-Qian Liu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China; Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jun-Li Zhang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Zhi-Fen Pan
- The Tuberculosis Division of the First Hospital of Jiaxing, Jiaxing, Zhejiang, China
| | - Jun-Tao Mai
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Heng-Jun Mei
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Yue Dai
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Lu Zhang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China; Shanghai Engineering Research Center of Industrial Microorganisms, Shanghai, China.
| | | |
Collapse
|
102
|
Burggraaf MJ, Speer A, Meijers AS, Ummels R, van der Sar AM, Korotkov KV, Bitter W, Kuijl C. Type VII Secretion Substrates of Pathogenic Mycobacteria Are Processed by a Surface Protease. mBio 2019; 10:e01951-19. [PMID: 31662454 PMCID: PMC6819658 DOI: 10.1128/mbio.01951-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/30/2019] [Indexed: 02/04/2023] Open
Abstract
Tuberculosis, one of the world's most severe infectious diseases, is caused by Mycobacterium tuberculosis A major weapon of this pathogen is a unique cell wall that protects the pathogen from eradication by the immune system. Mycobacteria have specialized secretion systems, e.g., type VII secretion or ESX systems, to transport substrates across this cell wall. The largest group of proteins that are secreted by these ESX systems are the PE proteins. Previously, it was shown that the N-terminal PE domain of about 100 amino acids is required for secretion. Here, we describe the identification of an aspartic protease, designated PecA, that removes (part of) this PE domain at the cell surface. Nearly all of the observed PE_PGRS proteins are processed by PecA. Interestingly, the protease itself is also a secreted PE protein and subject to self-cleavage. Furthermore, a defect in surface processing has no effect on the activity of the PE lipase protein LipY but does seem to affect the functioning of other virulence factors, as a pecA mutant strain of Mycobacterium marinum shows moderate attenuation in zebrafish larvae. In conclusion, our results reveal the presence of a functional aspartic acid protease in M. marinum that cleaves LipY, itself as well as other members of the PE_PGRS family. Finally, mutants lacking PecA show growth attenuation in vivo, suggesting that PecA plays a role during infection.IMPORTANCE Aspartic proteases are common in eukaryotes and retroviruses but are relatively rare among bacteria (N. D. Rawlings and A. Bateman, BMC Genomics 10:437, 2009, https://doi.org/10.1186/1471-2164-10-437). In contrast to eukaryotic aspartic proteases, bacterial aspartic proteases are generally located in the cytoplasm. We have identified a surface-associated mycobacterial aspartic protease, PecA, which cleaves itself and many other type VII secretion substrates of the PE_PGRS family. PecA is present in most pathogenic mycobacterial species, including M. tuberculosis In addition, pathogenicity of M. marinum is reduced in the ΔpecA mutant, indicating that PecA contributes to virulence.
Collapse
Affiliation(s)
- Maroeska J Burggraaf
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Alexander Speer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
| | - Aniek S Meijers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
| | - Roy Ummels
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
| | - Astrid M van der Sar
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
| | - Konstantin V Korotkov
- Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Wilbert Bitter
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
- Molecular Microbiology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Coenraad Kuijl
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam, Netherlands
| |
Collapse
|
103
|
Raze D, Segers J, Mille C, Slupek S, Lecher S, Coutte L, Antoine R, Ducrocq L, Rouanet C, Appelmelk BJ, Locht C. Coordinate regulation of virulence and metabolic genes by the transcription factor HbhR in Mycobacterium marinum. Mol Microbiol 2019; 113:52-67. [PMID: 31587365 DOI: 10.1111/mmi.14400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2019] [Indexed: 12/17/2022]
Abstract
The heparin-binding hemagglutinin (HBHA) is a multifunctional protein involved in adherence of Mycobacterium tuberculosis to non-phagocytic cells and in the formation of intracytosolic lipid inclusions. We demonstrate that the expression of hbhA is regulated by a transcriptional repressor, named HbhR, in Mycobacterium marinum. The hbhR gene, located upstream of hbhA, was identified by screening a transposon insertion library and detailed analysis of a mutant overproducing HBHA. HbhR was found to repress both hbhA and hbhR transcription by binding to the promoter regions of both genes. Complementation restored production of HBHA. RNA-seq analyses comparing the mutant and parental strains uncovered 27 genes, including hbhA, that were repressed and 20 genes activated by HbhR. Among the former, the entire locus of genes coding for a type-VII secretion system, including esxA, esxB and pe-ppe paralogs, as well as the gene coding for PspA, present in intracellular lipid vesicles, was identified, as was katG, a gene involved in the sensitivity to isoniazid. The latter category contains genes that play a role in diverse functions, such as metabolism and resistance to oxidative conditions. Thus, HbhR appears to be a master regulator, linking the transcriptional regulation of virulence, metabolic and antibiotic sensitivity genes in M. marinum.
Collapse
Affiliation(s)
- Dominique Raze
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Jérôme Segers
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Céline Mille
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Stéphanie Slupek
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Sophie Lecher
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Loïc Coutte
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Rudy Antoine
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Lucie Ducrocq
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Carine Rouanet
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| | - Ben J Appelmelk
- Molecular Microbiology/Medical Microbiology and Infection Control, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Camille Locht
- CIIL - Center for Infection and Immunity of Lille, Univ. Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 8204, Lille, F-59000, France
| |
Collapse
|
104
|
Aspatwar A, Kairys V, Rala S, Parikka M, Bozdag M, Carta F, Supuran CT, Parkkila S. Mycobacterium tuberculosis β-Carbonic Anhydrases: Novel Targets for Developing Antituberculosis Drugs. Int J Mol Sci 2019; 20:ijms20205153. [PMID: 31627429 PMCID: PMC6834203 DOI: 10.3390/ijms20205153] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022] Open
Abstract
The genome of Mycobacterium tuberculosis (Mtb) encodes three β-carbonic anhydrases (CAs, EC 4.2.1.1) that are crucial for the life cycle of the bacterium. The Mtbβ-CAs have been cloned and characterized, and the catalytic activities of the enzymes have been studied. The crystal structures of two of the enzymes have been resolved. In vitro inhibition studies have been conducted using different classes of carbonic anhydrase inhibitors (CAIs). In vivo inhibition studies of pathogenic bacteria containing β-CAs showed that β-CA inhibitors effectively inhibited the growth of pathogenic bacteria. The in vitro and in vivo studies clearly demonstrated that β-CAs of not only mycobacterial species, but also other pathogenic bacteria, can be targeted for developing novel antimycobacterial agents for treating tuberculosis and other microbial infections that are resistant to existing drugs. In this review, we present the molecular and structural data on three β-CAs of Mtb that will give us better insights into the roles of these enzymes in pathogenic bacterial species. We also present data from both in vitro inhibition studies using different classes of chemical compounds and in vivo inhibition studies focusing on M. marinum, a model organism and close relative of Mtb.
Collapse
Affiliation(s)
- Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland.
| | - Visvaldas Kairys
- Department of Bioinformatics, Institute of Biotechnology, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania.
| | - Sangeetha Rala
- Tampere University of Applied Sciences, Kuntokatu 3, FI-33520 Tampere, Finland.
| | - Mataleena Parikka
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland.
| | - Murat Bozdag
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via U. Schiff 6, I-50019 Sesto Fiorentino, Firenze, Italy.
| | - Fabrizio Carta
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via U. Schiff 6, I-50019 Sesto Fiorentino, Firenze, Italy.
| | - Claudiu T Supuran
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Via U. Schiff 6, I-50019 Sesto Fiorentino, Firenze, Italy.
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland.
- Fimlab Ltd. and Tampere University Hospital, FI-33520 Tampere, Finland.
| |
Collapse
|
105
|
Chavarro-Portillo B, Soto CY, Guerrero MI. Mycobacterium leprae's evolution and environmental adaptation. Acta Trop 2019; 197:105041. [PMID: 31152726 DOI: 10.1016/j.actatropica.2019.105041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 11/24/2022]
Abstract
Leprosy is an ancient disease caused by the acid-fast bacillus Mycobacterium leprae, also known as Hansen's bacillus. M. leprae is an obligate intracellular microorganism with a marked Schwann cell tropism and is the only human pathogen capable of invading the superficial peripheral nerves. The transmission mechanism of M. leprae is not fully understood; however, the nasal mucosa is accepted as main route of M. leprae entry to the human host. The complete sequencing and the comparative genome analysis show that M. leprae underwent a genome reductive evolution process, as result of lifestyle change and adaptation to different environments; some of lost genes are homologous to those of host cells. Thus, M. leprae reduced its genome size to 3.3 Mbp, contributing to obtain the lowest GC content (approximately 58%) among mycobacteria. The M. leprae genome contains 1614 open reading frames coding for functional proteins, and 1310 pseudogenes corresponding to 41% of the genome, approximately. Comparative analyses to different microorganisms showed that M. leprae possesses the highest content of pseudogenes among pathogenic and non-pathogenic bacteria and archaea. The pathogen adaptation into host cells, as the Schwann cells, brought about the reduction of the genome and induced multiple gene inactivation. The present review highlights the characteristics of genome's reductive evolution that M. leprae experiences in the genetic aspects compared with other pathogens. The possible mechanisms of pseudogenes formation are discussed.
Collapse
|
106
|
Madacki J, Mas Fiol G, Brosch R. Update on the virulence factors of the obligate pathogen Mycobacterium tuberculosis and related tuberculosis-causing mycobacteria. INFECTION GENETICS AND EVOLUTION 2019; 72:67-77. [DOI: 10.1016/j.meegid.2018.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/02/2018] [Accepted: 12/07/2018] [Indexed: 12/21/2022]
|
107
|
Kim BJ, Cha GY, Kim BR, Kook YH, Kim BJ. Insights From the Genome Sequence of Mycobacterium paragordonae, a Potential Novel Live Vaccine for Preventing Mycobacterial Infections: The Putative Role of Type VII Secretion Systems for an Intracellular Lifestyle Within Free-Living Environmental Predators. Front Microbiol 2019; 10:1524. [PMID: 31333625 PMCID: PMC6616192 DOI: 10.3389/fmicb.2019.01524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 06/18/2019] [Indexed: 12/31/2022] Open
Abstract
Mycobacterium paragordonae (Mpg) is a temperature-sensitive Mycobacterium species that can grow at permissive temperatures but fails to grow above 37°C. Due to this unique growth trait, Mpg has recently been proposed as a novel live vaccine candidate for the prevention of mycobacterial infections. Furthermore, the increasing frequency of the isolation of Mpg from water supply systems led us to hypothesize that the free-living amoeba system is the natural reservoir of Mpg. In this study, we report the complete 6.7-Mb genome sequence of Mpg and show that this genome comprises four different plasmids with lengths of 305 kb (pMpg-1), 144 kb (pMpg-2), 26 kb (pMpg-3), and 17 kb (pMpg-4). The first two plasmids, pMpg-1 and -2, encode distinct Type VII secretion systems (T7SS), ESX-P5 and ESX-2, respectively. Genome-based phylogeny indicated that Mpg is the closest relative to M. gordonae, which has a 7.7-Mb genome; phylogenetic analysis revealed an average of 86.68% nucleotide identity between these two species. The most important feature of Mpg genome is the acquisition of massive genes related to T7SS, which may have had effect on adaptation to their intracellular lifestyle within free-living environmental predators, such as amoeba. Comparisons of the resistance to bacterial killing within amoeba indicated that Mpg exhibited stronger resistance to amoeba killing compared to M. gordonae and M. marinum, further supporting our genome-based findings indicating the special adaptation of Mpg to free-living amoeba. We also determined that, among the strains studied, there were more shared CDS between M. tuberculosis and Mpg. In addition, the presence of diverse T7SSs in the Mpg genome, including an intact ESX-1, may suggest the feasibility of Mpg as a novel tuberculosis vaccine. Our data highlight a significant role of lateral gene transfer in the evolution of mycobacteria for niche diversification and for increasing the intracellular survival capacity.
Collapse
Affiliation(s)
- Byoung-Jun Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute, Institute of Endemic Diseases, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga-Yeong Cha
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute, Institute of Endemic Diseases, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute, Institute of Endemic Diseases, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon-Hoh Kook
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute, Institute of Endemic Diseases, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute, Institute of Endemic Diseases, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
108
|
Hodgkinson JW, Belosevic M, Elks PM, Barreda DR. Teleost contributions to the understanding of mycobacterial diseases. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 96:111-125. [PMID: 30776420 DOI: 10.1016/j.dci.2019.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 06/09/2023]
Abstract
Few pathogens have shaped human medicine as the mycobacteria. From understanding biological phenomena driving disease spread, to mechanisms of host-pathogen interactions and antibiotic resistance, the Mycobacterium genus continues to challenge and offer insights into the basis of health and disease. Teleost fish models of mycobacterial infections have progressed significantly over the past three decades, now supplying a range of unique tools and new opportunities to define the strategies employed by these Gram-positive bacteria to overcome host defenses, as well as those host antimicrobial pathways that can be used to limit its growth and spread. Herein, we take a comparative perspective and provide an update on the contributions of teleost models to our understanding of mycobacterial diseases.
Collapse
Affiliation(s)
- Jordan W Hodgkinson
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Philip M Elks
- The Bateson Centre, University of Sheffield, Western Bank, Sheffield, United Kingdom; Department of Infection and Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Daniel R Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
109
|
Ravid-Peretz S, Colorni A, Sharon G, Ucko M. Vaccination of European sea bass Dicentrarchus labrax with avirulent Mycobacterium marinum (iipA::kan mutant). FISH & SHELLFISH IMMUNOLOGY 2019; 90:317-327. [PMID: 31039442 DOI: 10.1016/j.fsi.2019.04.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 05/19/2023]
Abstract
Mycobacteriosis is a chronic progressive disease affecting teleost fishes all over the world. No vaccine is commercially available against its main etiological agent, Mycobacterium marinum. The mycobacterial gene responsible for invasion and intracellular persistence, iipA, is known to modulate M. marinum pathology. The innate and adaptive immune responses in sea bass (Dicentrarchus labrax) vaccinated with M. marinum iipA::kan mutant with (and without) the use of adjuvant, with (and without) a booster vaccination were monitored. The adjuvanted vaccine induced enhanced immune responses. TNF-α transcription levels were extremely high in spleen of the fish vaccinated with the addition of adjuvant in both fish vaccinated once and twice, followed by an IgM response highly specific for M. marinum. Also, histologically, granulomas started appearing in spleen and head-kidney tissues (but with no visible bacteria) within a month after vaccination, mainly with the adjuvanted vaccine. This was followed by reduction in pathology, as demonstrated by the lower number of granulomas (with visible bacteria), indicating that even heat-killed bacteria were able to elicit granulomatous formations. Adhesion of the internal organs and moderate pigmentation were observed in the perivisceral adipose tissue of nearly all vaccinated fish. Although the adjuvanted heat-killed avirulent iipA::kan mutant clearly induced a strong humoral and adaptive immune response, the booster treatment did not seem to have produced a significantly higher degree of protection from the disease compared to fish that received a single vaccination.
Collapse
Affiliation(s)
- Shay Ravid-Peretz
- Israel Oceanographic and Limnological Research, National Center for Mariculture, Eilat, 8811201, Israel
| | - Angelo Colorni
- Israel Oceanographic and Limnological Research, National Center for Mariculture, Eilat, 8811201, Israel
| | - Galit Sharon
- Israel Oceanographic and Limnological Research, National Center for Mariculture, Eilat, 8811201, Israel
| | - Michal Ucko
- Israel Oceanographic and Limnological Research, National Center for Mariculture, Eilat, 8811201, Israel.
| |
Collapse
|
110
|
Mashabela GT, de Wet TJ, Warner DF. Mycobacterium tuberculosis Metabolism. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0067-2019. [PMID: 31350832 PMCID: PMC10957194 DOI: 10.1128/microbiolspec.gpp3-0067-2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis is the cause of tuberculosis (TB), a disease which continues to overwhelm health systems in endemic regions despite the existence of effective combination chemotherapy and the widespread use of a neonatal anti-TB vaccine. For a professional pathogen, M. tuberculosis retains a surprisingly large proportion of the metabolic repertoire found in nonpathogenic mycobacteria with very different lifestyles. Moreover, evidence that additional functions were acquired during the early evolution of the M. tuberculosis complex suggests the organism has adapted (and augmented) the metabolic pathways of its environmental ancestor to persistence and propagation within its obligate human host. A better understanding of M. tuberculosis pathogenicity, however, requires the elucidation of metabolic functions under disease-relevant conditions, a challenge complicated by limited knowledge of the microenvironments occupied and nutrients accessed by bacilli during host infection, as well as the reliance in experimental mycobacteriology on a restricted number of experimental models with variable relevance to clinical disease. Here, we consider M. tuberculosis metabolism within the framework of an intimate host-pathogen coevolution. Focusing on recent advances in our understanding of mycobacterial metabolic function, we highlight unusual adaptations or departures from the better-characterized model intracellular pathogens. We also discuss the impact of these mycobacterial "innovations" on the susceptibility of M. tuberculosis to existing and experimental anti-TB drugs, as well as strategies for targeting metabolic pathways. Finally, we offer some perspectives on the key gaps in the current knowledge of fundamental mycobacterial metabolism and the lessons which might be learned from other systems.
Collapse
Affiliation(s)
- Gabriel T Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Current address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch, South Africa
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, South Africa
| |
Collapse
|
111
|
A New ESX-1 Substrate in Mycobacterium marinum That Is Required for Hemolysis but Not Host Cell Lysis. J Bacteriol 2019; 201:JB.00760-18. [PMID: 30833360 DOI: 10.1128/jb.00760-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
The ESX-1 (ESAT-6 system 1) secretion system plays a conserved role in the virulence of diverse mycobacterial pathogens, including the human pathogen Mycobacterium tuberculosis and M. marinum, an environmental mycobacterial species. The ESX-1 system promotes the secretion of protein virulence factors to the extracytoplasmic environment. The secretion of these proteins triggers the host response by lysing the phagosome during macrophage infection. Using proteomic analyses of the M. marinum secretome in the presence and absence of a functional ESX-1 system, we and others have hypothesized that MMAR_2894, a PE family protein, is a potential ESX-1 substrate in M. marinum We used genetic and quantitative proteomic approaches to determine if MMAR_2894 is secreted by the ESX-1 system, and we defined the requirement of MMAR_2894 for ESX-1-mediated secretion and virulence. We show that MMAR_2894 is secreted by the ESX-1 system in M. marinum and is itself required for the optimal secretion of the known ESX-1 substrates in M. marinum Moreover, we found that MMAR_2894 was differentially required for hemolysis and cytolysis of macrophages, two lytic activities ascribed to the M. marinum ESX-1 system.IMPORTANCE Both Mycobacterium tuberculosis, the cause of human tuberculosis (TB), and Mycobacterium marinum, a pathogen of ectotherms, use the ESX-1 secretion system to cause disease. There are many established similarities between the ESX-1 systems in M. tuberculosis and in M. marinum Yet the two bacteria infect different hosts, hinting at species-specific functions of the ESX-1 system. Our findings demonstrate that MMAR_2894 is a PE protein secreted by the ESX-1 system of M. marinum We show that MMAR_2894 is required for the optimal secretion of mycobacterial proteins required for disease. Because the MMAR_2894 gene is not conserved in M. tuberculosis, our findings demonstrate that MMAR_2894 may contribute to a species-specific function of the ESX-1 system in M. marinum, providing new insight into how the M. marinum and M. tuberculosis systems differ.
Collapse
|
112
|
Fieweger RA, Wilburn KM, VanderVen BC. Comparing the Metabolic Capabilities of Bacteria in the Mycobacterium tuberculosis Complex. Microorganisms 2019; 7:E177. [PMID: 31216777 PMCID: PMC6617402 DOI: 10.3390/microorganisms7060177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 02/06/2023] Open
Abstract
Pathogenic mycobacteria are known for their ability to maintain persistent infections in various mammals. The canonical pathogen in this genus is Mycobacterium tuberculosis and this bacterium is particularly successful at surviving and replicating within macrophages. Here, we will highlight the metabolic processes that M. tuberculosis employs during infection in macrophages and compare these findings with what is understood for other pathogens in the M. tuberculosis complex.
Collapse
Affiliation(s)
- Rachael A Fieweger
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| | - Kaley M Wilburn
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| | - Brian C VanderVen
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
113
|
Seth-Smith HM, Casanova C, Sommerstein R, Meinel DM, Abdelbary MM, Blanc DS, Droz S, Führer U, Lienhard R, Lang C, Dubuis O, Schlegel M, Widmer A, Keller PM, Marschall J, Egli A. Phenotypic and Genomic Analyses of Burkholderia stabilisClinical Contamination, Switzerland. Emerg Infect Dis 2019. [DOI: 10.3201/eid2406.172119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
114
|
Santucci P, Smichi N, Diomandé S, Poncin I, Point V, Gaussier H, Cavalier J, Kremer L, Canaan S. Dissecting the membrane lipid binding properties and lipase activity ofMycobacterium tuberculosisLipY domains. FEBS J 2019; 286:3164-3181. [DOI: 10.1111/febs.14864] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/25/2019] [Accepted: 04/25/2019] [Indexed: 12/29/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Laurent Kremer
- Institut de Recherche en Infectiologie de Montpellier (IRIM) CNRS UMR9004 Université de Montpellier France
- INSERM IRIM Montpellier France
| | | |
Collapse
|
115
|
Tandon H, Sharma A, Wadhwa S, Varadarajan R, Singh R, Srinivasan N, Sandhya S. Bioinformatic and mutational studies of related toxin-antitoxin pairs in Mycobacterium tuberculosis predict and identify key functional residues. J Biol Chem 2019; 294:9048-9063. [PMID: 31018964 DOI: 10.1074/jbc.ra118.006814] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/23/2019] [Indexed: 12/17/2022] Open
Abstract
Mycobacterium tuberculosis possesses an unusually large representation of type II toxin-antitoxin (TA) systems, whose functions and targets are mostly unknown. To better understand the basis of their unique expansion and to probe putative functional similarities among these systems, here we computationally and experimentally investigated their sequence relationships. Bioinformatic and phylogenetic investigations revealed that 51 sequences of the VapBC toxin family group into paralogous sub-clusters. On the basis of conserved sequence fingerprints within paralogues, we predicted functional residues and residues at the putative TA interface that are useful to evaluate TA interactions. Substitution of these likely functional residues abolished the toxin's growth-inhibitory activity. Furthermore, conducting similarity searches in 101 mycobacterial and ∼4500 other prokaryotic genomes, we assessed the relative conservation of the M. tuberculosis TA systems and found that most TA orthologues are well-conserved among the members of the M. tuberculosis complex, which cause tuberculosis in animal hosts. We found that soil-inhabiting, free-living Actinobacteria also harbor as many as 12 TA pairs. Finally, we identified five novel putative TA modules in M. tuberculosis. For one of them, we demonstrate that overexpression of the putative toxin, Rv2514c, induces bacteriostasis and that co-expression of the cognate antitoxin Rv2515c restores bacterial growth. Taken together, our findings reveal that toxin sequences are more closely related than antitoxin sequences in M. tuberculosis Furthermore, the identification of additional TA systems reported here expands the known repertoire of TA systems in M. tuberculosis.
Collapse
Affiliation(s)
- Himani Tandon
- From the Molecular Biophysics Unit, Indian Institute of Science, Bangalore-560012 and
| | - Arun Sharma
- the Tuberculosis Research Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, P. O. Box 4, Faridabad, Haryana-121001, India
| | - Saruchi Wadhwa
- the Tuberculosis Research Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, P. O. Box 4, Faridabad, Haryana-121001, India
| | - Raghavan Varadarajan
- From the Molecular Biophysics Unit, Indian Institute of Science, Bangalore-560012 and
| | - Ramandeep Singh
- the Tuberculosis Research Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, P. O. Box 4, Faridabad, Haryana-121001, India
| | | | - Sankaran Sandhya
- From the Molecular Biophysics Unit, Indian Institute of Science, Bangalore-560012 and
| |
Collapse
|
116
|
Behra PRK, Das S, Pettersson BMF, Shirreff L, DuCote T, Jacobsson KG, Ennis DG, Kirsebom LA. Extended insight into the Mycobacterium chelonae-abscessus complex through whole genome sequencing of Mycobacterium salmoniphilum outbreak and Mycobacterium salmoniphilum-like strains. Sci Rep 2019; 9:4603. [PMID: 30872669 PMCID: PMC6418233 DOI: 10.1038/s41598-019-40922-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/26/2019] [Indexed: 12/12/2022] Open
Abstract
Members of the Mycobacterium chelonae-abscessus complex (MCAC) are close to the mycobacterial ancestor and includes both human, animal and fish pathogens. We present the genomes of 14 members of this complex: the complete genomes of Mycobacterium salmoniphilum and Mycobacterium chelonae type strains, seven M. salmoniphilum isolates, and five M. salmoniphilum-like strains including strains isolated during an outbreak in an animal facility at Uppsala University. Average nucleotide identity (ANI) analysis and core gene phylogeny revealed that the M. salmoniphilum-like strains are variants of the human pathogen Mycobacterium franklinii and phylogenetically close to Mycobacterium abscessus. Our data further suggested that M. salmoniphilum separates into three branches named group I, II and III with the M. salmoniphilum type strain belonging to group II. Among predicted virulence factors, the presence of phospholipase C (plcC), which is a major virulence factor that makes M. abscessus highly cytotoxic to mouse macrophages, and that M. franklinii originally was isolated from infected humans make it plausible that the outbreak in the animal facility was caused by a M. salmoniphilum-like strain. Interestingly, M. salmoniphilum-like was isolated from tap water suggesting that it can be present in the environment. Moreover, we predicted the presence of mutational hotspots in the M. salmoniphilum isolates and 26% of these hotspots overlap with genes categorized as having roles in virulence, disease and defense. We also provide data about key genes involved in transcription and translation such as sigma factor, ribosomal protein and tRNA genes.
Collapse
Affiliation(s)
- Phani Rama Krishna Behra
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Sarbashis Das
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - B M Fredrik Pettersson
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Lisa Shirreff
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Tanner DuCote
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | | | - Don G Ennis
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Leif A Kirsebom
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden.
| |
Collapse
|
117
|
Child SA, Flint KL, Bruning JB, Bell SG. The characterisation of two members of the cytochrome P450 CYP150 family: CYP150A5 and CYP150A6 from Mycobacterium marinum. Biochim Biophys Acta Gen Subj 2019; 1863:925-934. [PMID: 30826435 DOI: 10.1016/j.bbagen.2019.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 02/27/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Actinobacteria, including the Mycobacteria, have a large component of cytochrome P450 family monooxygenases. This includes Mycobacterium tuberculosis, M. ulcerans and M. marinum, and M. vanbaalenii. These enzymes can abstract CH bonds and have important roles in natural product biosynthesis. METHODS Two members of the bacterial CYP150 family, CYP150A5 and CYP150A6 from M. marinum, were produced, purified and characterised. The potential substrate ranges of both enzymes were analysed and the monooxygenase activity of CYP150A5 was reconstituted using a physiological electron transfer partner system. CYP150A6 was structurally characterised by X-ray crystallography. RESULTS CYP150A5 was shown to bind various norisoprenoids and terpenoids. It could regioselectively hydroxylate β-ionol. The X-ray crystal structure of substrate-free CYP150A6 was solved to 1.5 Å. This displayed an open conformation with short F and G helices, an unresolved F-G loop region and exposed active site pocket. The active site residues could be identified and important variations were found among the CYP150A enzymes. Haem-binding azole inhibitors were identified for both enzymes. CONCLUSIONS The structure of CYP150A6 will facilitate the identification of physiological substrates and the design of better inhibitors for members of this P450 family. Based on the observed differences in substrate binding preference and sequence variations among the active site residues, their roles are predicted to be different. GENERAL SIGNIFICANCE Multiple CYP150 family members were found in many bacteria and are prevalent in the Mycobacteria including several human pathogens. Inhibition and structural data are reported here for these enzymes for the first time.
Collapse
Affiliation(s)
- Stella A Child
- Department of Chemistry, University of Adelaide, SA 5005, Australia
| | - Kate L Flint
- Department of Chemistry, University of Adelaide, SA 5005, Australia
| | - John B Bruning
- School of Biological Sciences, University of Adelaide, SA 5005, Australia
| | - Stephen G Bell
- Department of Chemistry, University of Adelaide, SA 5005, Australia.
| |
Collapse
|
118
|
Faber-Hammond JJ, Coyle KP, Bacheller SK, Roberts CG, Mellies JL, Roberts RB, Renn SCP. The intestinal environment as an evolutionary adaptation to mouthbrooding in the Astatotilapia burtoni cichlid. FEMS Microbiol Ecol 2019; 95:5315751. [PMID: 30753545 DOI: 10.1093/femsec/fiz016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Many of the various parental care strategies displayed by animals are accompanied by a significant reduction in food intake that imposes a substantial energy trade-off. Mouthbrooding, as seen in several species of fish in which the parent holds the developing eggs and fry in the buccal cavity, represents an extreme example of reduced food intake during parental investment and is accompanied by a range of physiological adaptations. In this study we use 16S sequencing to characterize the gut microbiota of female Astatotilapia burtoni cichlid fish throughout the obligatory phase of self-induced starvation during the brooding cycle in comparison to stage-matched females that have been denied food for the same duration. In addition to a reduction of gut epithelial turnover, we find a dramatic reduction in species diversity in brooding stages that recovers upon release of fry and refeeding that is not seen in females that are simply starved. Based on overall species diversity as well as differential abundance of specific bacterial taxa, we suggest that rather than reflecting a simple deprivation of caloric intake, the gut microbiota is more strongly influenced by physiological changes specific to mouthbrooding including the reduced epithelial turnover and possible production of antimicrobial agents.
Collapse
Affiliation(s)
| | - Kaitlin P Coyle
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, 3510 Thomas Hall, 112 Derieux Place, North Carolina State University, Raleigh, NC, USA
| | | | | | - Jay L Mellies
- Department of Biology, Reed College, Portland, Oregon, USA
| | - Reade B Roberts
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, 3510 Thomas Hall, 112 Derieux Place, North Carolina State University, Raleigh, NC, USA
| | - Suzy C P Renn
- Department of Biology, Reed College, Portland, Oregon, USA
| |
Collapse
|
119
|
van Wyk R, van Wyk M, Mashele SS, Nelson DR, Syed K. Comprehensive Comparative Analysis of Cholesterol Catabolic Genes/Proteins in Mycobacterial Species. Int J Mol Sci 2019; 20:ijms20051032. [PMID: 30818787 PMCID: PMC6429209 DOI: 10.3390/ijms20051032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
In dealing with Mycobacterium tuberculosis, the causative agent of the deadliest human disease—tuberculosis (TB)—utilization of cholesterol as a carbon source indicates the possibility of using cholesterol catabolic genes/proteins as novel drug targets. However, studies on cholesterol catabolism in mycobacterial species are scarce, and the number of mycobacterial species utilizing cholesterol as a carbon source is unknown. The availability of a large number of mycobacterial species’ genomic data affords an opportunity to explore and predict mycobacterial species’ ability to utilize cholesterol employing in silico methods. In this study, comprehensive comparative analysis of cholesterol catabolic genes/proteins in 93 mycobacterial species was achieved by deducing a comprehensive cholesterol catabolic pathway, developing a software tool for extracting homologous protein data and using protein structure and functional data. Based on the presence of cholesterol catabolic homologous proteins proven or predicted to be either essential or specifically required for the growth of M. tuberculosis H37Rv on cholesterol, we predict that among 93 mycobacterial species, 51 species will be able to utilize cholesterol as a carbon source. This study’s predictions need further experimental validation and the results should be taken as a source of information on cholesterol catabolism and genes/proteins involved in this process among mycobacterial species.
Collapse
Affiliation(s)
- Rochelle van Wyk
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, Free State, South Africa.
| | - Mari van Wyk
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, Free State, South Africa.
| | - Samson Sitheni Mashele
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, Free State, South Africa.
| | - David R Nelson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Khajamohiddin Syed
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa 3886, South Africa.
| |
Collapse
|
120
|
Child SA, Rossi VP, Bell SG. Selective ϖ-1 oxidation of fatty acids by CYP147G1 from Mycobacterium marinum. Biochim Biophys Acta Gen Subj 2019; 1863:408-417. [DOI: 10.1016/j.bbagen.2018.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/17/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
|
121
|
Bhandari S, Biswas S, Chaudhary A, Dutta S, Suguna K. Dodecameric structure of a small heat shock protein from Mycobacterium marinum M. Proteins 2019; 87:365-379. [PMID: 30632633 DOI: 10.1002/prot.25657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/24/2018] [Accepted: 01/05/2019] [Indexed: 12/31/2022]
Abstract
Small heat shock proteins (sHSPs) are ATP-independent molecular chaperones present ubiquitously in all kingdoms of life. Their low molecular weight subunits associate to form higher order structures. Under conditions of stress, sHSPs prevent aggregation of substrate proteins by undergoing rapid changes in their conformation or stoichiometry. Polydispersity and dynamic nature of these proteins have made structural investigations through crystallography a daunting task. In pathogens like Mycobacteria, sHSPs are immuno-dominant antigens, enabling survival of the pathogen within the host and contributing to disease persistence. We characterized sHSPs from Mycobacterium marinum M and determined the crystal structure of one of these. The protein crystallized in three different conditions as dodecamers, with dimers arranged in a tetrahedral fashion to form a closed cage-like architecture. Interestingly, we found a pentapeptide bound to the dodecamers revealing one of the modes of sHSP-substrate interaction. Further, we have observed that ATP inhibits the chaperoning activity of the protein.
Collapse
Affiliation(s)
- Spraha Bhandari
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Sreeparna Biswas
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Anuradha Chaudhary
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Somnath Dutta
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Kaza Suguna
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| |
Collapse
|
122
|
Santucci P, Dedaki C, Athanasoulis A, Gallorini L, Munoz A, Canaan S, Cavalier J, Magrioti V. Synthesis of Long‐Chain β‐Lactones and Their Antibacterial Activities against Pathogenic Mycobacteria. ChemMedChem 2019; 14:349-358. [DOI: 10.1002/cmdc.201800720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Pierre Santucci
- Aix-Marseille UniversitéCNRS, LISM, IMM FR3479 Marseille France
| | - Christina Dedaki
- Department of ChemistryNational and Kapodistrian University of Athens, Panepistimiopolis Athens 15771 Greece
| | - Alexandros Athanasoulis
- Department of ChemistryNational and Kapodistrian University of Athens, Panepistimiopolis Athens 15771 Greece
| | - Laura Gallorini
- Aix-Marseille UniversitéCNRS, LISM, IMM FR3479 Marseille France
| | - Anaïs Munoz
- Aix-Marseille UniversitéCNRS, LISM, IMM FR3479 Marseille France
| | - Stéphane Canaan
- Aix-Marseille UniversitéCNRS, LISM, IMM FR3479 Marseille France
| | | | - Victoria Magrioti
- Department of ChemistryNational and Kapodistrian University of Athens, Panepistimiopolis Athens 15771 Greece
| |
Collapse
|
123
|
Mycobacterium tuberculosis virulence inhibitors discovered by Mycobacterium marinum high-throughput screening. Sci Rep 2019; 9:26. [PMID: 30631100 PMCID: PMC6328581 DOI: 10.1038/s41598-018-37176-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 12/03/2018] [Indexed: 01/08/2023] Open
Abstract
High-throughput screening facilities do not generally support biosafety level 3 organisms such as Mycobacterium tuberculosis. To discover not only antibacterials, but also virulence inhibitors with either bacterial or host cell targets, an assay monitoring lung fibroblast survival upon infection was developed and optimized for 384-plate format and robotic liquid handling. By using Mycobacterium marinum as surrogate organism, 28,000 compounds were screened at biosafety level 2 classification, resulting in 49 primary hits. Exclusion of substances with unfavourable properties and known antimicrobials resulted in 11 validated hits of which 7 had virulence inhibiting properties and one had bactericidal effect also in wild type Mycobacterium tuberculosis. This strategy to discover virulence inhibitors using a model organism in high-throughput screening can be a valuable tool for other researchers working on drug discovery against tuberculosis and other biosafety level 3 infectious agents.
Collapse
|
124
|
Seth-Smith HMB, Imkamp F, Tagini F, Cuénod A, Hömke R, Jahn K, Tschacher A, Grendelmeier P, Bättig V, Erb S, Reinhard M, Rütimann G, Borrell S, Gagneux S, Casanova C, Droz S, Osthoff M, Tamm M, Nübel U, Greub G, Keller PM, Egli A. Discovery and Characterization of Mycobacterium basiliense sp. nov., a Nontuberculous Mycobacterium Isolated From Human Lungs. Front Microbiol 2019; 9:3184. [PMID: 30671031 PMCID: PMC6331445 DOI: 10.3389/fmicb.2018.03184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022] Open
Abstract
Bacteria belonging to the genus Mycobacterium are predominantly responsible for pulmonary diseases; most notably Mycobacterium tuberculosis causes granulomatous pulmonary infections. Here we describe a novel slow growing mycobacterial species isolated from respiratory samples from five patients, four with underlying pulmonary disease. The isolates were characterized by biochemical and molecular techniques, including whole genome sequencing. Biochemical characteristics generally match those of M. marinum and M. ulcerans; however, the most striking difference of the new species is its ability to grow at 37°C. The new species was found to grow in human macrophages, but not amoebae, suggesting a pathogenic rather than an environmental lifestyle. Phylogenetic analysis reveals a deep-rooting relationship to M. marinum and M. ulcerans. A complete genome sequence was obtained through combining short and long-read sequencing, providing a genome of 5.6 Mb. The genome appears to be highly intact, syntenic with that of M. marinum, with very few insertion sequences. A vast array of virulence factors includes 283 PE/PPE surface-associated proteins, making up 10% of the coding capacity, and 22 non-ribosomal peptide synthase clusters. A comparison of six clinical isolates from the five patients shows that they differ by up to two single nucleotide polymorphisms, suggesting a common source of infection. Our findings are in accordance with the recognition of a new taxonomic entity. We propose the name M. basiliense, as all isolates were found in patients from the Basel area of Switzerland.
Collapse
Affiliation(s)
- Helena M. B. Seth-Smith
- Division of Clinical Microbiology, University Hospital Basel, Basel, Switzerland
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Frank Imkamp
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Florian Tagini
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Aline Cuénod
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Rico Hömke
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- National Center for Mycobacteria, Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Kathleen Jahn
- Division of Pneumology, University Hospital Basel, Basel, Switzerland
| | - Anne Tschacher
- Division of Pneumology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Peter Grendelmeier
- Division of Pneumology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Veronika Bättig
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Erb
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Miriam Reinhard
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Sonia Borrell
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Sebastien Gagneux
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Carlo Casanova
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Sara Droz
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Michael Osthoff
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Internal Medicine, University Hospital Basel, Basel, Switzerland
| | - Michael Tamm
- Division of Pneumology, University Hospital Basel, Basel, Switzerland
| | - Ulrich Nübel
- Leibniz Institute DSMZ, German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Gilbert Greub
- Institute of Microbiology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Peter M. Keller
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
- National Center for Mycobacteria, Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Adrian Egli
- Division of Clinical Microbiology, University Hospital Basel, Basel, Switzerland
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
125
|
Abstract
Tuberculosis (TB) remains a leading cause of death globally among infectious diseases that has killed more numbers of people than any other infectious diseases. Animal models have become the lynchpin for mimicking human infectious diseases. Research on TB could be facilitated by animal challenge models such as the guinea pig, mice, rabbit and non-human primates. No single model presents all aspects of disease pathogenesis due to considerable differences in disease resistance/susceptibility between these models. Availability of a wide range of animal strains, Mycobacterium tuberculosis strains, route of infection and doses affect the disease progression and intervention outcome. Different animal models have contributed significantly to the drug and vaccine development, identification of biomarkers, understanding of TB immunopathogenesis and host genetic influence on infection. In this review, the commonly used animal models in TB research are discussed along with their advantages and limitations.
Collapse
Affiliation(s)
- Amit Kumar Singh
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| | - Umesh D Gupta
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| |
Collapse
|
126
|
Johansen MD, Hortle E, Kasparian JA, Romero A, Novoa B, Figueras A, Britton WJ, de Silva K, Purdie AC, Oehlers SH. Analysis of mycobacterial infection-induced changes to host lipid metabolism in a zebrafish infection model reveals a conserved role for LDLR in infection susceptibility. FISH & SHELLFISH IMMUNOLOGY 2018; 83:238-242. [PMID: 30219383 DOI: 10.1016/j.fsi.2018.09.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/20/2018] [Accepted: 09/12/2018] [Indexed: 06/08/2023]
Abstract
Changes to lipid metabolism are well-characterised consequences of human tuberculosis infection but their functional relevance are not clearly elucidated in these or other host-mycobacterial systems. The zebrafish-Mycobacterium marinum infection model is used extensively to model many aspects of human-M. tuberculosis pathogenesis but has not been widely used to study the role of infection-induced lipid metabolism. We find mammalian mycobacterial infection-induced alterations in host Low Density Lipoprotein metabolism are conserved in the zebrafish model of mycobacterial pathogenesis. Depletion of LDLR, a key lipid metabolism node, decreased M. marinum burden, and corrected infection-induced altered lipid metabolism resulting in decreased LDL and reduced the rate of macrophage transformation into foam cells. Our results demonstrate a conserved role for infection-induced alterations to host lipid metabolism, and specifically the LDL-LDLR axis, across host-mycobacterial species pairings.
Collapse
Affiliation(s)
- Matt D Johansen
- Tuberculosis Research Program Centenary Institute, Sydney Medical School The University of Sydney, Camperdown, NSW, Australia; Sydney School of Veterinary Science and Marie Bashir Institute The University of Sydney, Camden, NSW, Australia
| | - Elinor Hortle
- Tuberculosis Research Program Centenary Institute, Sydney Medical School The University of Sydney, Camperdown, NSW, Australia
| | - Joshua A Kasparian
- Tuberculosis Research Program Centenary Institute, Sydney Medical School The University of Sydney, Camperdown, NSW, Australia; Sydney School of Veterinary Science and Marie Bashir Institute The University of Sydney, Camden, NSW, Australia
| | - Alejandro Romero
- Institute of Marine Research (IIM), Spanish National Research Council (CSIC), Vigo, Spain
| | - Beatriz Novoa
- Institute of Marine Research (IIM), Spanish National Research Council (CSIC), Vigo, Spain
| | - Antonio Figueras
- Institute of Marine Research (IIM), Spanish National Research Council (CSIC), Vigo, Spain
| | - Warwick J Britton
- Tuberculosis Research Program Centenary Institute, Sydney Medical School The University of Sydney, Camperdown, NSW, Australia; Tuberculosis Research Program Centenary Institute, Sydney Medical School and Marie Bashir Institute The University of Sydney, Camperdown, NSW, Australia
| | - Kumudika de Silva
- Sydney School of Veterinary Science and Marie Bashir Institute The University of Sydney, Camden, NSW, Australia
| | - Auriol C Purdie
- Sydney School of Veterinary Science and Marie Bashir Institute The University of Sydney, Camden, NSW, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program Centenary Institute, Sydney Medical School The University of Sydney, Camperdown, NSW, Australia; Tuberculosis Research Program Centenary Institute, Sydney Medical School and Marie Bashir Institute The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
127
|
Abstract
Humans encounter mycobacterial species due to their ubiquity in different environmental niches. In many individuals, pathogenic mycobacterial species may breach our first-line barrier defenses of the innate immune system and modulate the activation of phagocytes to cause disease of the respiratory tract or the skin and soft tissues, sometimes resulting in disseminated infection. Cutaneous mycobacterial infections may cause a wide range of clinical manifestations, which are divided into four main disease categories: (i) cutaneous manifestations of Mycobacterium tuberculosis infection, (ii) Buruli ulcer caused by Mycobacterium ulcerans and other related slowly growing mycobacteria, (iii) leprosy caused by Mycobacterium leprae and Mycobacterium lepromatosis, and (iv) cutaneous infections caused by rapidly growing mycobacteria. Clinically, cutaneous mycobacterial infections present with widely different clinical presentations, including cellulitis, nonhealing ulcers, subacute or chronic nodular lesions, abscesses, superficial lymphadenitis, verrucous lesions, and other types of findings. Mycobacterial infections of the skin and subcutaneous tissue are associated with important stigma, deformity, and disability. Geography-based environmental exposures influence the epidemiology of cutaneous mycobacterial infections. Cutaneous tuberculosis exhibits different clinical phenotypes acquired through different routes, including via extrinsic inoculation of the tuberculous bacilli and dissemination to the skin from other sites, or represents hypersensitivity reactions to M. tuberculosis infection. In many settings, leprosy remains an important cause of neurological impairment, deformity, limb loss, and stigma. Mycobacterium lepromatosis, a mycobacterial species related to M. leprae, is linked to diffuse lepromatous leprosy of Lucio and Latapí. Mycobacterium ulcerans produces a mycolactone toxin that leads to subcutaneous tissue destruction and immunosuppression, resulting in deep ulcerations that often produce substantial disfigurement and disability. Mycobacterium marinum, a close relative of M. ulcerans, is an important cause of cutaneous sporotrichoid nodular lymphangitic lesions. Among patients with advanced immunosuppression, Mycobacterium kansasii, the Mycobacterium avium-intracellulare complex, and Mycobacterium haemophilum may cause cutaneous or disseminated disease. Rapidly growing mycobacteria, including the Mycobacterium abscessus group, Mycobacterium chelonei, and Mycobacterium fortuitum, are increasingly recognized pathogens in cutaneous infections associated particularly with plastic surgery and cosmetic procedures. Skin biopsies of cutaneous lesions to identify acid-fast staining bacilli and cultures represent the cornerstone of diagnosis. Additionally, histopathological evaluation of skin biopsy specimens may be useful in identifying leprosy, Buruli ulcer, and cutaneous tuberculosis. Molecular assays are useful in some cases. The treatment for cutaneous mycobacterial infections depends on the specific pathogen and therefore requires a careful consideration of antimicrobial choices based on official treatment guidelines.
Collapse
|
128
|
Aspatwar A, Winum JY, Carta F, Supuran CT, Hammaren M, Parikka M, Parkkila S. Carbonic Anhydrase Inhibitors as Novel Drugs against Mycobacterial β-Carbonic Anhydrases: An Update on In Vitro and In Vivo Studies. Molecules 2018; 23:molecules23112911. [PMID: 30413024 PMCID: PMC6278287 DOI: 10.3390/molecules23112911] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Mycobacteria cause a variety of diseases, such as tuberculosis, leprosy, and opportunistic diseases in immunocompromised people. The treatment of these diseases is problematic, necessitating the development of novel treatment strategies. Recently, β-carbonic anhydrases (β-CAs) have emerged as potential drug targets in mycobacteria. The genomes of mycobacteria encode for three β-CAs that have been cloned and characterized from Mycobacterium tuberculosis (Mtb) and the crystal structures of two of the enzymes have been determined. Different classes of inhibitor molecules against Mtb β-CAs have subsequently been designed and have been shown to inhibit these mycobacterial enzymes in vitro. The inhibition of these centrally important mycobacterial enzymes leads to reduced growth of mycobacteria, lower virulence, and impaired biofilm formation. Thus, the inhibition of β-CAs could be a novel approach for developing drugs against the severe diseases caused by pathogenic mycobacteria. In the present article, we review the data related to in vitro and in vivo inhibition studies in the field.
Collapse
Affiliation(s)
- Ashok Aspatwar
- Faculty of Medicine and Health Technology, University of Tampere, 33014 Tampere, Finland.
| | - Jean-Yves Winum
- Institut des Biomolécules Max Mousseron (IBMM) UMR 5247 CNRS, ENSCM, Université de Montpellier, 34296 Montpellier CEDEX 05, France.
| | - Fabrizio Carta
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (Firenze), Italy.
| | - Claudiu T Supuran
- Neurofarba Department, Sezione di Chimica Farmaceutica e Nutraceutica, Università degli Studi di Firenze, 50019 Sesto Fiorentino (Firenze), Italy.
| | - Milka Hammaren
- Faculty of Medicine and Health Technology, University of Tampere, 33014 Tampere, Finland.
| | - Mataleena Parikka
- Faculty of Medicine and Health Technology, University of Tampere, 33014 Tampere, Finland.
- Oral and Maxillofacial Unit, Tampere University Hospital, 33521 Tampere, Finland.
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, University of Tampere, 33014 Tampere, Finland.
- Fimlab Ltd. and Tampere University Hospital, 33520 Tampere, Finland.
| |
Collapse
|
129
|
Child SA, Bradley JM, Pukala TL, Svistunenko DA, Le Brun NE, Bell SG. Electron transfer ferredoxins with unusual cluster binding motifs support secondary metabolism in many bacteria. Chem Sci 2018; 9:7948-7957. [PMID: 30542550 PMCID: PMC6237146 DOI: 10.1039/c8sc01286e] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/21/2018] [Indexed: 01/06/2023] Open
Abstract
The proteins responsible for controlling electron transfer in bacterial secondary metabolism are not always known or characterised. Here we demonstrate that many bacteria contain a set of unfamiliar ferredoxin encoding genes which are associated with those of cytochrome P450 (CYP) monooxygenases and as such are involved in anabolic and catabolic metabolism. The model organism Mycobacterium marinum M contains eleven of these genes which encode [3Fe-4S] or [4Fe-4S] single cluster containing ferredoxins but which have unusual iron-sulfur cluster binding motif sequences, CXX?XXC(X) n CP, where '?' indicates a variable amino acid residue. Rather than a cysteine residue, which is highly conserved in [4Fe-4S] clusters, or alanine or glycine residues, which are common in [3Fe-4S] ferredoxins, these genes encode at this position histidine, asparagine, tyrosine, serine, threonine or phenylalanine. We have purified, characterised and reconstituted the activity of several of these CYP/electron transfer partner systems and show that all those examined contain a [3Fe-4S] cluster. Furthermore, the ferredoxin used and the identity of the variable motif residue in these proteins affects the functionality of the monooxygenase system and has a significant influence on the redox properties of the ferredoxins. Similar ferredoxin encoding genes were identified across Mycobacterium species, including in the pathogenic M. tuberculosis and M. ulcerans, as well as in a wide range of other bacteria such as Rhodococcus and Streptomyces. In the majority of instances these are associated with CYP genes. These ferredoxin systems are important in controlling electron transfer across bacterial secondary metabolite production processes which include antibiotic and pigment formation among others.
Collapse
Affiliation(s)
- Stella A Child
- Department of Chemistry , University of Adelaide , SA 5005 , Australia .
| | - Justin M Bradley
- Centre for Molecular and Structural Biochemistry , School of Chemistry , University of East Anglia , Norwich Research Park , Norwich , NR4 7TJ , UK
| | - Tara L Pukala
- Department of Chemistry , University of Adelaide , SA 5005 , Australia .
| | - Dimitri A Svistunenko
- School of Biological Sciences , University of Essex , Wivenhoe Park , Colchester CO4 3SQ , UK
| | - Nick E Le Brun
- Centre for Molecular and Structural Biochemistry , School of Chemistry , University of East Anglia , Norwich Research Park , Norwich , NR4 7TJ , UK
| | - Stephen G Bell
- Department of Chemistry , University of Adelaide , SA 5005 , Australia .
| |
Collapse
|
130
|
Luukinen H, Hammarén MM, Vanha-Aho LM, Parikka M. Modeling Tuberculosis in Mycobacterium marinum Infected Adult Zebrafish. J Vis Exp 2018. [PMID: 30346391 DOI: 10.3791/58299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis is currently the deadliest human pathogen causing 1.7 million deaths and 10.4 million infections every year. Exposure to this bacterium causes a wide disease spectrum in humans ranging from a sterilized infection to an actively progressing deadly disease. The most common form is the latent tuberculosis, which is asymptomatic, but has the potential to reactivate into a fulminant disease. Adult zebrafish and its natural pathogen Mycobacterium marinum have recently proven to be an applicable model to study the wide disease spectrum of tuberculosis. Importantly, spontaneous latency and reactivation as well as adaptive immune responses in the context of mycobacterial infection can be studied in this model. In this article, we describe methods for the experimental infection of adult zebrafish, the collection of internal organs for the extraction of nucleic acids for the measurement of mycobacterial loads and host immune responses by quantitative PCR. The in-house-developed, M. marinum-specific qPCR assay is more sensitive than the traditional plating methods as it also detects DNA from non-dividing, dormant or recently dead mycobacteria. As both DNA and RNA are extracted from the same individual, it is possible to study the relationships between the diseased state, and the host and pathogen gene-expression. The adult zebrafish model for tuberculosis thus presents itself as a highly applicable, non-mammalian in vivo system to study host-pathogen interactions.
Collapse
Affiliation(s)
- Hanna Luukinen
- Faculty of Medicine and Life Sciences, University of Tampere;
| | | | | | - Mataleena Parikka
- Faculty of Medicine and Life Sciences, University of Tampere; Oral and Maxillofacial Unit, Tampere University Hospital
| |
Collapse
|
131
|
A microfluidic cell-trapping device to study dynamic host-microbe interactions at the single-cell level. Methods Cell Biol 2018. [PMID: 30165958 DOI: 10.1016/bs.mcb.2018.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Single-cell imaging of host-microbe interactions over time is impeded by cellular motility because the cells under scrutiny tend to migrate out of the imaging field. To overcome this technical challenge, we developed a microfluidic platform for imaging hundreds of individual motile phagocytic cells and bacteria within microfluidic traps that restrict their movement. The interaction of trapped host cells and bacteria is monitored by long-term time-lapse microscopy, allowing direct visualization of all stages of infection at the single-cell level. The medium flowing through the microfluidic device can be changed quickly and precisely, permitting the real-time imaging of cellular responses to antibiotics or other environmental stresses. Here, we demonstrate the potential applications of this approach by co-culturing the phagocytic amoeba Dictyostelium discoideum with the intracellular pathogen Mycobacterium marinum. However, the platform can be adapted easily for use with other host cells or microorganisms. This approach will provide new insights into host-pathogen interactions that cannot be studied using conventional population-based assays.
Collapse
|
132
|
Thompson CR, Champion MM, Champion PA. Quantitative N-Terminal Footprinting of Pathogenic Mycobacteria Reveals Differential Protein Acetylation. J Proteome Res 2018; 17:3246-3258. [PMID: 30080413 DOI: 10.1021/acs.jproteome.8b00373] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
N-terminal acetylation (NTA) is a post-transcriptional modification of proteins that is conserved from bacteria to humans. In bacteria, the enzymes that mediate protein NTA also promote antimicrobial resistance. In pathogenic mycobacteria, which cause human tuberculosis and other chronic infections, NTA has been linked to pathogenesis and stress response, yet the fundamental biology underlying NTA of mycobacterial proteins remains unclear. We enriched, defined, and quantified the NT-acetylated populations of both cell-associated and secreted proteins from both the human pathogen, Mycobacterium tuberculosis, and the nontuberculous opportunistic pathogen, Mycobacterium marinum. We used a parallel N-terminal enrichment strategy from proteolytic digests coupled to charge-based selection and stable isotope ratio mass spectrometry. We show that NTA of the mycobacterial proteome is abundant, diverse, and primarily on Thr residues, which is unique compared with other bacteria. We isolated both the acetylated and unacetylated forms of 256 proteins, indicating that NTA of mycobacterial proteins is homeostatic. We identified 16 mycobacterial proteins with differential levels of NTA on the cytoplasmic and secreted forms, linking protein modification and localization. Our findings reveal novel biology underlying the NTA of mycobacterial proteins, which may provide a basis to understand NTA in mycobacterial physiology, pathogenesis, and antimicrobial resistance.
Collapse
|
133
|
Das S, Pettersson BMF, Behra PRK, Mallick A, Cheramie M, Ramesh M, Shirreff L, DuCote T, Dasgupta S, Ennis DG, Kirsebom LA. Extensive genomic diversity among Mycobacterium marinum strains revealed by whole genome sequencing. Sci Rep 2018; 8:12040. [PMID: 30104693 PMCID: PMC6089878 DOI: 10.1038/s41598-018-30152-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/25/2018] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium marinum is the causative agent for the tuberculosis-like disease mycobacteriosis in fish and skin lesions in humans. Ubiquitous in its geographical distribution, M. marinum is known to occupy diverse fish as hosts. However, information about its genomic diversity is limited. Here, we provide the genome sequences for 15 M. marinum strains isolated from infected humans and fish. Comparative genomic analysis of these and four available genomes of the M. marinum strains M, E11, MB2 and Europe reveal high genomic diversity among the strains, leading to the conclusion that M. marinum should be divided into two different clusters, the "M"- and the "Aronson"-type. We suggest that these two clusters should be considered to represent two M. marinum subspecies. Our data also show that the M. marinum pan-genome for both groups is open and expanding and we provide data showing high number of mutational hotspots in M. marinum relative to other mycobacteria such as Mycobacterium tuberculosis. This high genomic diversity might be related to the ability of M. marinum to occupy different ecological niches.
Collapse
Affiliation(s)
- Sarbashis Das
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - B M Fredrik Pettersson
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Phani Rama Krishna Behra
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Amrita Mallick
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Martin Cheramie
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Malavika Ramesh
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Lisa Shirreff
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Tanner DuCote
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Santanu Dasgupta
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden
| | - Don G Ennis
- Department of Biology, University of Louisiana, Lafayette, Louisiana, USA
| | - Leif A Kirsebom
- Department of Cell and Molecular Biology, Box 596, Biomedical Centre, SE-751 24, Uppsala, Sweden.
| |
Collapse
|
134
|
Mechanisms of Bacterial Tolerance and Persistence in the Gastrointestinal and Respiratory Environments. Clin Microbiol Rev 2018; 31:31/4/e00023-18. [PMID: 30068737 DOI: 10.1128/cmr.00023-18] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Pathogens that infect the gastrointestinal and respiratory tracts are subjected to intense pressure due to the environmental conditions of the surroundings. This pressure has led to the development of mechanisms of bacterial tolerance or persistence which enable microorganisms to survive in these locations. In this review, we analyze the general stress response (RpoS mediated), reactive oxygen species (ROS) tolerance, energy metabolism, drug efflux pumps, SOS response, quorum sensing (QS) bacterial communication, (p)ppGpp signaling, and toxin-antitoxin (TA) systems of pathogens, such as Escherichia coli, Salmonella spp., Vibrio spp., Helicobacter spp., Campylobacter jejuni, Enterococcus spp., Shigella spp., Yersinia spp., and Clostridium difficile, all of which inhabit the gastrointestinal tract. The following respiratory tract pathogens are also considered: Staphylococcus aureus, Pseudomonas aeruginosa, Acinetobacter baumannii, Burkholderia cenocepacia, and Mycobacterium tuberculosis Knowledge of the molecular mechanisms regulating the bacterial tolerance and persistence phenotypes is essential in the fight against multiresistant pathogens, as it will enable the identification of new targets for developing innovative anti-infective treatments.
Collapse
|
135
|
Harjula SKE, Ojanen MJT, Taavitsainen S, Nykter M, Rämet M. Interleukin 10 mutant zebrafish have an enhanced interferon gamma response and improved survival against a Mycobacterium marinum infection. Sci Rep 2018; 8:10360. [PMID: 29985419 PMCID: PMC6037744 DOI: 10.1038/s41598-018-28511-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 06/20/2018] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis ranks as one of the world’s deadliest infectious diseases causing more than a million casualties annually. IL10 inhibits the function of Th1 type cells, and IL10 deficiency has been associated with an improved resistance against Mycobacterium tuberculosis infection in a mouse model. Here, we utilized M. marinum infection in the zebrafish (Danio rerio) as a model for studying Il10 in the host response against mycobacteria. Unchallenged, nonsense il10e46/e46 mutant zebrafish were fertile and phenotypically normal. Following a chronic mycobacterial infection, il10e46/e46 mutants showed enhanced survival compared to the controls. This was associated with an increased expression of the Th cell marker cd4-1 and a shift towards a Th1 type immune response, which was demonstrated by the upregulated expression of tbx21 and ifng1, as well as the down-regulation of gata3. In addition, at 8 weeks post infection il10e46/e46 mutant zebrafish had reduced expression levels of proinflammatory cytokines tnfb and il1b, presumably indicating slower progress of the infection. Altogether, our data show that Il10 can weaken the immune defense against M. marinum infection in zebrafish by restricting ifng1 response. Importantly, our findings support the relevance of M. marinum infection in zebrafish as a model for tuberculosis.
Collapse
Affiliation(s)
- Sanna-Kaisa E Harjula
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Markus J T Ojanen
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Laboratory of Immunoregulation, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Sinja Taavitsainen
- Laboratory of Computational Biology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Matti Nykter
- Laboratory of Computational Biology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland. .,Department of Pediatrics, Tampere University Hospital, Tampere, Finland. .,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland. .,PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
136
|
Accelerating Early Antituberculosis Drug Discovery by Creating Mycobacterial Indicator Strains That Predict Mode of Action. Antimicrob Agents Chemother 2018; 62:AAC.00083-18. [PMID: 29661879 DOI: 10.1128/aac.00083-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
Due to the rise of drug-resistant forms of tuberculosis, there is an urgent need for novel antibiotics to effectively combat these cases and shorten treatment regimens. Recently, drug screens using whole-cell analyses have been shown to be successful. However, current high-throughput screens focus mostly on stricto sensu life/death screening that give little qualitative information. In doing so, promising compound scaffolds or nonoptimized compounds that fail to reach inhibitory concentrations are missed. To accelerate early tuberculosis (TB) drug discovery, we performed RNA sequencing on Mycobacterium tuberculosis and Mycobacterium marinum to map the stress responses that follow upon exposure to subinhibitory concentrations of antibiotics with known targets, ciprofloxacin, ethambutol, isoniazid, streptomycin, and rifampin. The resulting data set comprises the first overview of transcriptional stress responses of mycobacteria to different antibiotics. We show that antibiotics can be distinguished based on their specific transcriptional stress fingerprint. Notably, this fingerprint was more distinctive in M. marinum We decided to use this to our advantage and continue with this model organism. A selection of diverse antibiotic stress genes was used to construct stress reporters. In total, three functional reporters were constructed to respond to DNA damage, cell wall damage, and ribosomal inhibition. Subsequently, these reporter strains were used to screen a small anti-TB compound library to predict the mode of action. In doing so, we identified the putative modes of action for three novel compounds, which confirms the utility of our approach.
Collapse
|
137
|
Jiang H, Sun J, Chen Y, Chen Z, Wang L, Gao W, Shi Y, Zhang W, Mei Y, Chokkakula S, Vissa V, Jiang T, Wu A, Wang H. Landscape of the genome and host cell response of Mycobacterium shigaense reveals pathogenic features. Emerg Microbes Infect 2018; 7:112. [PMID: 29934568 PMCID: PMC6015043 DOI: 10.1038/s41426-018-0116-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 02/08/2023]
Abstract
A systems approach was used to explore the genome and transcriptome of Mycobacterium shigaense, a new opportunistic pathogen isolated from a patient with a skin infection, and the host response transcriptome was assessed using a macrophage infection model. The M. shigaense genome comprises 5,207,883 bp, with 67.2% G+C content and 5098 predicted coding genes. Evolutionarily, the bacterium belongs to a cluster in the phylogenetic tree along with three target opportunistic pathogenic strains, namely, M. avium, M. triplex and M. simiae. Potential virulence genes are indeed expressed by M. shigaense under culture conditions. Phenotypically, M. shigaense had similar infection and replication capacities in a macrophage model as the opportunistic species compared to M. tuberculosis. M. shigaense activated NF-κB, TNF, cytokines and chemokines in the host innate immune-related signaling pathways and elicited an early response shared with pathogenic bacilli except M. tuberculosis. M. shigaense upregulated specific host response genes such as TLR7, CCL4 and CXCL5. We performed an integrated and comparative analysis of M. shigaense. Multigroup comparison indicated certain differences with typical pathogenic bacilli in terms of gene features and the macrophage response.
Collapse
Affiliation(s)
- Haiqin Jiang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Jiya Sun
- Suzhou Institute of Systems Medicine, Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, 215021, China
| | - Yanqing Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Zhiming Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Le Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Wei Gao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Ying Shi
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Wenyue Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Youming Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Santosh Chokkakula
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Varalakshmi Vissa
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China
| | - Taijiao Jiang
- Suzhou Institute of Systems Medicine, Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, 215021, China.
| | - Aiping Wu
- Suzhou Institute of Systems Medicine, Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, 215021, China.
| | - Hongsheng Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China. .,Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China.
| |
Collapse
|
138
|
Complete Genome Sequence of Mycobacterium marinum ATCC 927 T, Obtained Using Nanopore and Illumina Sequencing Technologies. GENOME ANNOUNCEMENTS 2018; 6:6/20/e00397-18. [PMID: 29773624 PMCID: PMC5958268 DOI: 10.1128/genomea.00397-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mycobacterium marinum is a slowly growing, broad-host-range mycobacterial species. Here, we report the complete genome sequence of a Mycobacterium marinum type strain that was isolated from tubercles of diseased fish. This sequence will provide essential information for future taxonomic and comparative genome studies of its relatives.
Collapse
|
139
|
Esx Paralogs Are Functionally Equivalent to ESX-1 Proteins but Are Dispensable for Virulence in Mycobacterium marinum. J Bacteriol 2018; 200:JB.00726-17. [PMID: 29555701 DOI: 10.1128/jb.00726-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/11/2018] [Indexed: 12/20/2022] Open
Abstract
Mycobacterium marinum is a nontuberculous pathogen of poikilothermic fish and an opportunistic human pathogen. Like tuberculous mycobacteria, the M. marinum M strain requires the ESX-1 (ESAT-6 system 1) secretion system for virulence in host cells. EsxB and EsxA, two major virulence factors exported by the ESX-1 system, are encoded by the esxBA genes within the ESX-1 locus. Deletion of the esxBA genes abrogates ESX-1 export and attenuates M. marinum in ex vivo and in vivo models of infection. Interestingly, there are several duplications of the esxB and esxA genes (esxB_1, esxB_2, esxA_1, esxA_2, and esxA_3) in the M. marinum M genome located outside the ESX-1 locus. We sought to understand if this region, known as ESX-6, contributes to ESX-1-mediated virulence. We found that deletion of the esxB_1 gene alone or the entire ESX-6 locus did not impact ESX-1 export or function, supporting the idea that the esxBA genes present at the ESX-1 locus are the primary contributors to ESX-1-mediated virulence. Nevertheless, overexpression of the esxB_1 locus complemented ESX-1 function in the ΔesxBA strain, signifying that the two loci are functionally equivalent. Our findings raise questions about why duplicate versions of the esxBA genes are maintained in the M. marinum M genome and how these proteins, which are functionally equivalent to virulence factors, contribute to mycobacterial biology.IMPORTANCEMycobacterium tuberculosis is the causative agent of the human disease tuberculosis (TB). There are 10.4 million cases and 1.7 million TB-associated deaths annually, making TB a leading cause of death globally. Nontuberculous mycobacteria (NTM) cause chronic human infections that are acquired from the environment. Despite differences in disease etiology, both tuberculous and NTM pathogens use the ESX-1 secretion system to cause disease. The nontubercular mycobacterial species, Mycobacterium marinum, has additional copies of specific ESX-1 genes. Our findings demonstrate that the duplicated genes do not contribute to virulence but can substitute for virulence factors in M. marinum These findings suggest that the duplicated genes may play a specific role in NTM biology.
Collapse
|
140
|
A protocol for culturing environmental strains of the Buruli ulcer agent, Mycobacterium ulcerans. Sci Rep 2018; 8:6778. [PMID: 29712992 PMCID: PMC5928104 DOI: 10.1038/s41598-018-25278-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 04/13/2018] [Indexed: 11/08/2022] Open
Abstract
Contaminations and fastidiousness of M. ulcerans may have both hamper isolation of strains from environmental sources. We aimed to optimize decontamination and culture of environmental samples to circumvent both limitations. Three strains of M. ulcerans cultured onto Middlebrook 7H10 at 30 °C for 20 days yielded a significantly higher number of colonies in micro-aerophilic atmosphere compared to ambient atmosphere, 5% CO2 and anaerobic atmosphere. In a second step, we observed that M. ulcerans genome uniquely encoded chitinase, fucosidase and A-D-GlcNAc-diphosphoryl polyprenol A-3-L-rhamnosyl transferase giving M. ulcerans the potential to metabolize chitine, fucose and N-acetyl galactosamine (NAG), respectively. A significant growth-promoting effect of 0.2 mg/mL chitin (p < 0.05), 0.01 mg/mL N-acetyl galactosamine (p < 0.05), 0.01 mg/mL fucose (p < 0.05) was observed with M. ulcerans indicating that NAG alone or combined with fucose and chitin could complement Middlebrook 7H10. Finally, the protocol combining 1% chlorhexidine decontamination with micro-aerophilic incubation on Middlebrook 7H10 medium containing chitin (0.2%), NAG (0.01%) and fucose (0.01%) medium and auto-fluorescence detection of colonies allowed for the isolation of one mycolactone-encoding strain from Thryonomys swinderianus (aulacode) feces specimens collected near the Kossou Dam, Côte d'Ivoire. We propose that incubation of chlorhexidine-decontaminated environmental specimens on Middlebrook 7H10-enriched medium under micro-aerophilic atmosphere at 30 °C may be used for the tentative isolation of M. ulcerans strains from potential environmental sources.
Collapse
|
141
|
Parvez A, Giri S, Giri GR, Kumari M, Bisht R, Saxena P. Novel Type III Polyketide Synthases Biosynthesize Methylated Polyketides in Mycobacterium marinum. Sci Rep 2018; 8:6529. [PMID: 29695799 PMCID: PMC5916927 DOI: 10.1038/s41598-018-24980-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/12/2018] [Indexed: 01/09/2023] Open
Abstract
Mycobacterial pathogenesis is hallmarked by lipidic polyketides that decorate the cell envelope and mediate infection. However, factors mediating persistence remain largely unknown. Dynamic cell wall remodeling could facilitate the different pathogenic phases. Recent studies have implicated type III polyketide synthases (PKSs) in cell wall alterations in several bacteria. Comparative genome analysis revealed several type III pks gene clusters in mycobacteria. In this study, we report the functional characterization of two novel type III PKSs, MMAR_2470 and MMAR_2474, in Mycobacterium marinum. These type III pkss belong to a unique pks genomic cluster conserved exclusively in pathogenic mycobacteria. Cell-free reconstitution assays and high-resolution mass spectrometric analyses revealed methylated polyketide products in independent reactions of both proteins. MMAR_2474 protein exceptionally biosynthesized methylated alkyl-resorcinol and methylated acyl-phloroglucinol products from the same catalytic core. Structure-based homology modeling, product docking, and mutational studies identified residues that could facilitate the distinctive catalysis of these proteins. Functional investigations in heterologous mycobacterial strain implicated MMAR_2474 protein to be vital for mycobacterial survival in stationary biofilms. Our investigations provide new insights into type III PKSs conserved in pathogenic mycobacterial species.
Collapse
Affiliation(s)
- Amreesh Parvez
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India
| | - Samir Giri
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.,Department of Ecology, School of Biology, University of Osnabrück, Osnabrück, 49076, Germany
| | - Gorkha Raj Giri
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India
| | - Monika Kumari
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.,Department of Biochemistry, University College of Medical Sciences, Delhi, 110095, India
| | - Renu Bisht
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India
| | - Priti Saxena
- Chemical Biology Group, Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110021, India.
| |
Collapse
|
142
|
Rodriguez-Rivera FP, Zhou X, Theriot JA, Bertozzi CR. Acute Modulation of Mycobacterial Cell Envelope Biogenesis by Front-Line Tuberculosis Drugs. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Frances P. Rodriguez-Rivera
- Department of Chemistry; University of California; Berkeley CA 94720 USA
- Department of Chemistry; Stanford University; Stanford CA 94305 USA
| | - Xiaoxue Zhou
- Department of Biochemistry; Stanford University School of Medicine; Stanford CA 94305 USA
| | - Julie A. Theriot
- Department of Biochemistry; Stanford University School of Medicine; Stanford CA 94305 USA
- Department of Microbiology and Immunology; Stanford University School of Medicine; Stanford CA 94305 USA
- Howard Hughes Medical Institute; Stanford University; Stanford CA 94305 USA
| | - Carolyn R. Bertozzi
- Department of Chemistry; Stanford University; Stanford CA 94305 USA
- Howard Hughes Medical Institute; Stanford University; Stanford CA 94305 USA
| |
Collapse
|
143
|
Rodriguez-Rivera FP, Zhou X, Theriot JA, Bertozzi CR. Acute Modulation of Mycobacterial Cell Envelope Biogenesis by Front-Line Tuberculosis Drugs. Angew Chem Int Ed Engl 2018; 57:5267-5272. [PMID: 29392891 DOI: 10.1002/anie.201712020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/28/2018] [Indexed: 02/06/2023]
Abstract
Front-line tuberculosis (TB) drugs have been characterized extensively in vitro and in vivo with respect to gene expression and cell viability. However, little work has been devoted to understanding their effects on the physiology of the cell envelope, one of the main targets of this clinical regimen. Herein, we use metabolic labeling methods to visualize the effects of TB drugs on cell envelope dynamics in mycobacterial species. We developed a new fluorophore-trehalose conjugate to visualize trehalose monomycolates of the mycomembrane using super-resolution microscopy. We also probed the relationship between mycomembrane and peptidoglycan dynamics using a dual metabolic labeling strategy. Finally, we found that metabolic labeling of both cell envelope structures reports on drug effects on cell physiology in two hours, far faster than a genetic sensor of cell envelope stress. Our work provides insight into acute drug effects on cell envelope biogenesis in live mycobacteria.
Collapse
Affiliation(s)
- Frances P Rodriguez-Rivera
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA.,Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Xiaoxue Zhou
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
144
|
Myllymäki H, Niskanen M, Luukinen H, Parikka M, Rämet M. Identification of protective postexposure mycobacterial vaccine antigens using an immunosuppression-based reactivation model in the zebrafish. Dis Model Mech 2018; 11:11/3/dmm033175. [PMID: 29590635 PMCID: PMC5897733 DOI: 10.1242/dmm.033175] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/14/2018] [Indexed: 12/28/2022] Open
Abstract
Roughly one third of the human population carries a latent Mycobacterium tuberculosis infection, with a 5-10% lifetime risk of reactivation to active tuberculosis and further spreading the disease. The mechanisms leading to the reactivation of a latent Mycobacterium tuberculosis infection are insufficiently understood. Here, we used a natural fish pathogen, Mycobacterium marinum, to model the reactivation of a mycobacterial infection in the adult zebrafish (Danio rerio). A low-dose intraperitoneal injection (∼40 colony-forming units) led to a latent infection, with mycobacteria found in well-organized granulomas surrounded by a thick layer of fibrous tissue. A latent infection could be reactivated by oral dexamethasone treatment, which led to disruption of the granuloma structures and dissemination of bacteria. This was associated with the depletion of lymphocytes, especially CD4+ T cells. Using this model, we verified that ethambutol is effective against an active disease but not a latent infection. In addition, we screened 15 mycobacterial antigens as postexposure DNA vaccines, of which RpfB and MMAR_4207 reduced bacterial burdens upon reactivation, as did the Ag85-ESAT-6 combination. In conclusion, the adult zebrafish-M. marinum infection model provides a feasible tool for examining the mechanisms of reactivation in mycobacterial infections, and for screening vaccine and drug candidates.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Henna Myllymäki
- BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere FI-33014, Finland
| | - Mirja Niskanen
- BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere FI-33014, Finland
| | - Hanna Luukinen
- BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere FI-33014, Finland
| | - Mataleena Parikka
- BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere FI-33014, Finland.,Oral and Maxillofacial Unit, Tampere University Hospital, Tampere FI-33521, Finland
| | - Mika Rämet
- BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere FI-33014, Finland.,Department of Pediatrics, Tampere University Hospital, Tampere FI-33521, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu FI-90220, Finland.,PEDEGO Research Unit, and, Medical Research Center, University of Oulu, Oulu FI-90014, Finland
| |
Collapse
|
145
|
|
146
|
Structural and functional characterisation of the cytochrome P450 enzyme CYP268A2 from Mycobacterium marinum. Biochem J 2018; 475:705-722. [DOI: 10.1042/bcj20170946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 11/17/2022]
Abstract
Members of the cytochrome P450 monooxygenase family CYP268 are found across a broad range of Mycobacterium species including the pathogens Mycobacterium avium, M. colombiense, M. kansasii, and M. marinum. CYP268A2, from M. marinum, which is the first member of this family to be studied, was purified and characterised. CYP268A2 was found to bind a variety of substrates with high affinity, including branched and straight chain fatty acids (C10–C12), acetate esters, and aromatic compounds. The enzyme was also found to bind phenylimidazole inhibitors but not larger azoles, such as ketoconazole. The monooxygenase activity of CYP268A2 was efficiently reconstituted using heterologous electron transfer partner proteins. CYP268A2 hydroxylated geranyl acetate and trans-pseudoionone at a terminal methyl group to yield (2E,6E)-8-hydroxy-3,7-dimethylocta-2,6-dien-1-yl acetate and (3E,5E,9E)-11-hydroxy-6,10-dimethylundeca-3,5,9-trien-2-one, respectively. The X-ray crystal structure of CYP268A2 was solved to a resolution of 2.0 Å with trans-pseudoionone bound in the active site. The overall structure was similar to that of the related phytanic acid monooxygenase CYP124A1 enzyme from Mycobacterium tuberculosis, which shares 41% sequence identity. The active site is predominantly hydrophobic, but includes the Ser99 and Gln209 residues which form hydrogen bonds with the terminal carbonyl group of the pseudoionone. The structure provided an explanation on why CYP268A2 shows a preference for shorter substrates over the longer chain fatty acids which bind to CYP124A1 and the selective nature of the catalysed monooxygenase activity.
Collapse
|
147
|
Entwistle FM, Coote PJ. Evaluation of greater wax moth larvae, Galleria mellonella, as a novel in vivo model for non-tuberculosis Mycobacteria infections and antibiotic treatments. J Med Microbiol 2018; 67:585-597. [PMID: 29458557 DOI: 10.1099/jmm.0.000696] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
PURPOSE To evaluate the suitability of Galleria mellonella larvae as an in vivo model and drug-screening tool for mycobacteria infections. METHODOLOGY Larvae were infected using a range of inoculum sizes from a variety of rapid-growing mycobacteria, including strains of M. fortuitum, M. marinum and M. aurum. Larval survival, internal bacterial burden and the effects of amikacin, ciprofloxacin, ethambutol, isoniazid and rifampicin treatment on larval survival were measured over 144 h. The effects of these anti-mycobacterial drugs on phagocytosis and circulating haemocyte numbers were also examined using microscopy. RESULTS Larval survival decreased after infection with M. fortuitum and M. marinum in a dose-dependent manner, but remained unaffected by M. aurum. Heat-killed bacteria did not cause larval death. Where antibiotic monotherapy was efficacious, larval survival post-infection increased in a dose-dependent fashion. However, efficacy varied between different antibiotics and species of infecting mycobacteria and, apart from rifampicin, efficacy in vivo correlated poorly with the in vitro minimum inhibitory concentrations (MICs). Combinations of antibiotics led to higher survival of infected larvae than antibiotic monotherapy. Selected antibiotic treatments that enhanced larval survival reduced the overall internal burden of infecting mycobacteria, but did not eradicate the pathogens. Administration of amikacin or ethambutol to uninfected larvae induced an initial transient increase in the numbers of circulating haemocytes and reduced the phagocytic rate of haemocytes in larvae infected with M. marinum. CONCLUSIONS This report demonstrates the potential of employing a wax moth larvae model for studying fast-growing mycobacteria infections, and as a cheap, effective system for initial screening of novel treatments.
Collapse
Affiliation(s)
- Frances M Entwistle
- Biomedical Sciences Research Complex, School of Biology, The North Haugh, University of St Andrews, Fife, KY16 9ST, UK
| | - Peter J Coote
- Biomedical Sciences Research Complex, School of Biology, The North Haugh, University of St Andrews, Fife, KY16 9ST, UK
| |
Collapse
|
148
|
Abstract
Mycobacteria are a major human health problem globally. Regarding tuberculosis the situation is worsened by the poor efficacy of current vaccine regimens and by emergence of drug-resistant strains (Manjelievskaia J et al, Trans R Soc Trop Med Hyg 110: 110, 2016; Pereira et al., Lancet Infect Dis 12:300-306, 2012; http://www.who.int/tb/publications/global_report/en/) undermining both disease-prevention and available treatments. Thus, increased basic understanding of mycobacterial-and particularly Mycobacterium tuberculosis-virulence strategies and pathogenesis is of great importance. To this end several in vivo infection models are available (Guirado and Schlesinger, Front Immunol 4:98, 2013; Leung et al., Eur J Immunol 43:2246-2254, 2013; Patel et al., J Lab Physicians 3:75-79, 2011; van Leeuwen et al., Cold Spring Harb Perspect Med 5:a018580, 2015). While these models all have their merits they also exhibit limitations, and none perfectly mimics all aspects of human tuberculosis. Thus, there is a need for multiple models that may complement each other, ultimately allowing us to gain true insight into the pathogenesis of mycobacterial infections.Here, we describe a recently developed mouse model of Mycobacterium marinum infection that allows kinetic and quantitative studies of disease progression in live animals [8]. Notably, this model exhibits features of human tuberculosis not replicated in M. tuberculosis infected mice, and may provide an important complement to the field. For example, granulomas in the M. marinum model develop central caseating necrosis (Carlsson et al., PLoS Pathog 6:e1000895, 2010), a hallmark of granulomas in human tuberculosis normally not replicated in murine M. tuberculosis infection. Moreover, while tuberculosis is heterogeneous and presents with a continuum of active and latent disease, M. tuberculosis infected mice essentially lack this dynamic range and do not replicate latency (Guirado and Schlesinger, Front Immunol 4:98, 2013; Patel et al., J Lab Physicians 3(2):75-79, 2011). In contrast, M. marinum infected mice may naturally develop latency, as suggested by reduced inflammation and healing of the diseased tissue while low numbers of bacteria persist in granulomatous lesions (Carlsson et al., PLoS Pathog 6:e1000895, 2010). Thus, infection with M. marinum may offer a unique murine model for studying granuloma formation as well as latency-and possibly also for studies of disease-reactivation. In addition to the in vivo model, we describe infection of bone marrow-derived murine macrophages, an in vitro platform enabling detailed mechanistic studies of host-pathogen interactions occurring in the principal host target cell for pathogenic mycobacteria.
Collapse
|
149
|
Cardenal-Muñoz E, Barisch C, Lefrançois LH, López-Jiménez AT, Soldati T. When Dicty Met Myco, a (Not So) Romantic Story about One Amoeba and Its Intracellular Pathogen. Front Cell Infect Microbiol 2018; 7:529. [PMID: 29376033 PMCID: PMC5767268 DOI: 10.3389/fcimb.2017.00529] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/18/2017] [Indexed: 01/06/2023] Open
Abstract
In recent years, Dictyostelium discoideum has become an important model organism to study the cell biology of professional phagocytes. This amoeba not only shares many molecular features with mammalian macrophages, but most of its fundamental signal transduction pathways are conserved in humans. The broad range of existing genetic and biochemical tools, together with its suitability for cell culture and live microscopy, make D. discoideum an ideal and versatile laboratory organism. In this review, we focus on the use of D. discoideum as a phagocyte model for the study of mycobacterial infections, in particular Mycobacterium marinum. We look in detail at the intracellular cycle of M. marinum, from its uptake by D. discoideum to its active or passive egress into the extracellular medium. In addition, we describe the molecular mechanisms that both the mycobacterial invader and the amoeboid host have developed to fight against each other, and compare and contrast with those developed by mammalian phagocytes. Finally, we introduce the methods and specific tools that have been used so far to monitor the D. discoideum-M. marinum interaction.
Collapse
Affiliation(s)
- Elena Cardenal-Muñoz
- Department of Biochemistry, Sciences II, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | | | | | | | | |
Collapse
|
150
|
Buruli Ulcer, a Prototype for Ecosystem-Related Infection, Caused by Mycobacterium ulcerans. Clin Microbiol Rev 2017; 31:31/1/e00045-17. [PMID: 29237707 DOI: 10.1128/cmr.00045-17] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Buruli ulcer is a noncontagious disabling cutaneous and subcutaneous mycobacteriosis reported by 33 countries in Africa, Asia, Oceania, and South America. The causative agent, Mycobacterium ulcerans, derives from Mycobacterium marinum by genomic reduction and acquisition of a plasmid-borne, nonribosomal cytotoxin mycolactone, the major virulence factor. M. ulcerans-specific sequences have been readily detected in aquatic environments in food chains involving small mammals. Skin contamination combined with any type of puncture, including insect bites, is the most plausible route of transmission, and skin temperature of <30°C significantly correlates with the topography of lesions. After 30 years of emergence and increasing prevalence between 1970 and 2010, mainly in Africa, factors related to ongoing decreasing prevalence in the same countries remain unexplained. Rapid diagnosis, including laboratory confirmation at the point of care, is mandatory in order to reduce delays in effective treatment. Parenteral and potentially toxic streptomycin-rifampin is to be replaced by oral clarithromycin or fluoroquinolone combined with rifampin. In the absence of proven effective primary prevention, avoiding skin contamination by means of clothing can be implemented in areas of endemicity. Buruli ulcer is a prototype of ecosystem pathology, illustrating the impact of human activities on the environment as a source for emerging tropical infectious diseases.
Collapse
|