1
|
Faozia S, Hossain T, Cho KH. The Dlt and LiaFSR systems derepress SpeB production independently in the Δpde2 mutant of Streptococcus pyogenes. Front Cell Infect Microbiol 2023; 13:1293095. [PMID: 38029265 PMCID: PMC10679467 DOI: 10.3389/fcimb.2023.1293095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
The second messenger molecule, c-di-AMP, plays a critical role in pathogenesis and virulence in S. pyogenes. We previously reported that deleting the c-di-AMP phosphodiesterase gene pde2 severely suppresses SpeB production at the transcriptional level. We performed transposon mutagenesis to gain insight into the mechanism of how Pde2 is involved in SpeB regulation. We identified one of the genes of the dlt operon, dltX, as a suppressor of the SpeB-null phenotype of the Δpde2 mutant. The dlt operon consists of five genes, dltX, dltA, dltB, dltC, and dltD in many Gram-positive bacteria, and its function is to incorporate D-alanine into lipoteichoic acids. DltX, a small membrane protein, is a newly identified member of the operon. The in-frame deletion of dltX or insertional inactivation of dltA in the Δpde2 mutant restored SpeB production, indicating that D-alanylation is crucial for the suppressor phenotype. These mutations did not affect the growth in lab media but showed increased negative cell surface charge and enhanced sensitivity to polymyxin B. Considering that dlt mutations change cell surface charge and sensitivity to cationic antimicrobial peptides, we examined the LiaFSR system that senses and responds to cell envelope stress. The ΔliaR mutation in the Δpde2 mutant also derepressed SpeB production, like the ΔdltX mutation. LiaFSR controls speB expression by regulating the expression of the transcriptional regulator SpxA2. However, the Dlt system did not regulate spxA2 expression. The SpeB phenotype of the Δpde2ΔdltX mutant in higher salt media differed from that of the Δpde2ΔliaR mutant, suggesting a unique pathway for the Dlt system in SpeB production, possibly related to ion transport or turgor pressure regulation.
Collapse
Affiliation(s)
| | | | - Kyu Hong Cho
- Department of Biology, Indiana State University, Terre Haute, IN, United States
| |
Collapse
|
2
|
Host–Pathogen Interactions of Marine Gram-Positive Bacteria. BIOLOGY 2022; 11:biology11091316. [PMID: 36138795 PMCID: PMC9495620 DOI: 10.3390/biology11091316] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Complex interactions between marine Gram-positive pathogens and fish hosts in the marine environment can result in diseases of economically important finfish, which cause economic losses in the aquaculture industry. Understanding how these pathogens interact with the fish host and generate disease will contribute to efficient prophylactic measures and treatments. To our knowledge, there are no systematic reviews on marine Gram-positive pathogens. Therefore, here we reviewed the host–pathogen interactions of marine Gram-positive pathogens from the pathogen-centric and host-centric points of view. Abstract Marine Gram-positive bacterial pathogens, including Renibacterium salmoninarum, Mycobacterium marinum, Nocardia seriolae, Lactococcus garvieae, and Streptococcus spp. cause economic losses in marine fish aquaculture worldwide. Comprehensive information on these pathogens and their dynamic interactions with their respective fish–host systems are critical to developing effective prophylactic measures and treatments. While much is known about bacterial virulence and fish immune response, it is necessary to synthesize the knowledge in terms of host–pathogen interactions as a centerpiece to establish a crucial connection between the intricate details of marine Gram-positive pathogens and their fish hosts. Therefore, this review provides a holistic view and discusses the different stages of the host–pathogen interactions of marine Gram-positive pathogens. Gram-positive pathogens can invade fish tissues, evade the fish defenses, proliferate in the host system, and modulate the fish immune response. Marine Gram-positive pathogens have a unique set of virulence factors that facilitate adhesion (e.g., adhesins, hemagglutination activity, sortase, and capsules), invasion (e.g., toxins, hemolysins/cytolysins, the type VII secretion system, and immune-suppressive proteins), evasion (e.g., free radical quenching, actin-based motility, and the inhibition of phagolysosomal fusion), and proliferation and survival (e.g., heme utilization and siderophore-mediated iron acquisition systems) in the fish host. After infection, the fish host initiates specific innate and adaptive immune responses according to the extracellular or intracellular mechanism of infection. Although efforts have continued to be made in understanding the complex interplay at the host–pathogen interface, integrated omics-based investigations targeting host–pathogen–marine environment interactions hold promise for future research.
Collapse
|
3
|
Indraratna AD, Everest-Dass A, Skropeta D, Sanderson-Smith M. OUP accepted manuscript. FEMS Microbiol Rev 2022; 46:6519265. [PMID: 35104861 PMCID: PMC9075583 DOI: 10.1093/femsre/fuac001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/29/2021] [Accepted: 01/25/2022] [Indexed: 11/12/2022] Open
Abstract
Host carbohydrates, or glycans, have been implicated in the pathogenesis of many bacterial infections. Group A Streptococcus (GAS) is a Gram-positive bacterium that readily colonises the skin and oropharynx, and is a significant cause of mortality in humans. While the glycointeractions orchestrated by many other pathogens are increasingly well-described, the understanding of the role of human glycans in GAS disease remains incomplete. Although basic investigation into the mechanisms of GAS disease is ongoing, several glycointeractions have been identified and are examined herein. The majority of research in this context has focussed on bacterial adherence, however, glycointeractions have also been implicated in carbohydrate metabolism; evasion of host immunity; biofilm adaptations; and toxin-mediated haemolysis. The involvement of human glycans in these diverse avenues of pathogenesis highlights the clinical value of understanding glycointeractions in combatting GAS disease.
Collapse
Affiliation(s)
- Anuk D Indraratna
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Arun Everest-Dass
- Institute for Glycomics, Griffith University, Gold Coast Campus, Parklands Drive, Southport, Queensland, 4215, Australia
| | - Danielle Skropeta
- Illawarra Health and Medical Research Institute, Northfields Ave, Keiraville New South Wales 2522, Australia
- School of Chemistry and Molecular Bioscience, Molecular Horizons, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia
| | - Martina Sanderson-Smith
- Corresponding author: Illawarra Health and Medical Research Institute, Bld 32, University of Wollongong, Northfields Avenue, Keiraville, New South Wales, 2522, Australia. Tel: +61 2 42981935; E-mail:
| |
Collapse
|
4
|
c-di-AMP-Regulated K + Importer KtrAB Affects Biofilm Formation, Stress Response, and SpeB Expression in Streptococcus pyogenes. Infect Immun 2021; 89:IAI.00317-20. [PMID: 33468578 DOI: 10.1128/iai.00317-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/14/2021] [Indexed: 11/20/2022] Open
Abstract
The second messenger cyclic di-AMP (c-di-AMP) controls biofilm formation, stress response, and virulence in Streptococcus pyogenes The deletion of the c-di-AMP synthase gene, dacA, results in pleiotropic effects including reduced expression of the secreted protease SpeB. Here, we report a role for K+ transport in c-di-AMP-mediated SpeB expression. The deletion of ktrB in the ΔdacA mutant restores SpeB expression. KtrB is a subunit of the K+ transport system KtrAB that forms a putative high-affinity K+ importer. KtrB forms a membrane K+ channel, and KtrA acts as a cytosolic gating protein that controls the transport capacity of the system by binding ligands including c-di-AMP. SpeB induction in the ΔdacA mutant by K+ specific ionophore treatment also supports the importance of cellular K+ balance in SpeB production. The ΔdacA ΔktrB double deletion mutant not only produces wild-type levels of SpeB but also partially or fully reverts the defective ΔdacA phenotypes of biofilm formation and stress responses, suggesting that many ΔdacA phenotypes are due to cellular K+ imbalance. However, the null pathogenicity of the ΔdacA mutant in a murine subcutaneous infection model is not restored by ktrB deletion, suggesting that c-di-AMP controls not only cellular K+ balance but also other metabolic and/or virulence pathways. The deletion of other putative K+ importer genes, kup and kimA, does not phenocopy the deletion of ktrB regarding SpeB induction in the ΔdacA mutant, suggesting that KtrAB is the primary K+ importer that is responsible for controlling cellular K+ levels under laboratory growth conditions.
Collapse
|
5
|
Cornax I, Zulk J, Olson J, Fulde M, Nizet V, Patras KA. Novel Models of Streptococcus canis Colonization and Disease Reveal Modest Contributions of M-Like (SCM) Protein. Microorganisms 2021; 9:microorganisms9010183. [PMID: 33467030 PMCID: PMC7829700 DOI: 10.3390/microorganisms9010183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/14/2021] [Indexed: 12/16/2022] Open
Abstract
Streptococcus canis is a common colonizing bacterium of the urogenital tract of cats and dogs that can also cause invasive disease in these animal populations and in humans. Although the virulence mechanisms of S. canis are not well-characterized, an M-like protein, SCM, has recently identified been as a potential virulence factor. SCM is a surface-associated protein that binds to host plasminogen and IgGs suggesting its possible importance in host-pathogen interactions. In this study, we developed in vitro and ex vivo blood component models and murine models of S. canis vaginal colonization, systemic infection, and dermal infection to compare the virulence potential of the zoonotic S. canis vaginal isolate G361 and its isogenic SCM-deficient mutant (G361∆scm). We found that while S. canis establishes vaginal colonization and causes invasive disease in vivo, the contribution of the SCM protein to virulence phenotypes in these models is modest. We conclude that SCM is dispensable for invasive disease in murine models and for resistance to human blood components ex vivo, but may contribute to mucosal persistence, highlighting a potential contribution to the recently appreciated genetic diversity of SCM across strains and hosts.
Collapse
Affiliation(s)
- Ingrid Cornax
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (I.C.); (J.O.); (V.N.)
| | - Jacob Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA;
| | - Joshua Olson
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (I.C.); (J.O.); (V.N.)
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Victor Nizet
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA; (I.C.); (J.O.); (V.N.)
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA;
- Correspondence:
| |
Collapse
|
6
|
Alves-Barroco C, Paquete-Ferreira J, Santos-Silva T, Fernandes AR. Singularities of Pyogenic Streptococcal Biofilms - From Formation to Health Implication. Front Microbiol 2021; 11:584947. [PMID: 33424785 PMCID: PMC7785724 DOI: 10.3389/fmicb.2020.584947] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023] Open
Abstract
Biofilms are generally defined as communities of cells involved in a self-produced extracellular matrix adhered to a surface. In biofilms, the bacteria are less sensitive to host defense mechanisms and antimicrobial agents, due to multiple strategies, that involve modulation of gene expression, controlled metabolic rate, intercellular communication, composition, and 3D architecture of the extracellular matrix. These factors play a key role in streptococci pathogenesis, contributing to therapy failure and promoting persistent infections. The species of the pyogenic group together with Streptococcus pneumoniae are the major pathogens belonging the genus Streptococcus, and its biofilm growth has been investigated, but insights in the genetic origin of biofilm formation are limited. This review summarizes pyogenic streptococci biofilms with details on constitution, formation, and virulence factors associated with formation.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - João Paquete-Ferreira
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Teresa Santos-Silva
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| |
Collapse
|
7
|
Vyas HKN, Proctor EJ, McArthur J, Gorman J, Sanderson-Smith M. Current Understanding of Group A Streptococcal Biofilms. Curr Drug Targets 2020; 20:982-993. [PMID: 30947646 PMCID: PMC6700754 DOI: 10.2174/1389450120666190405095712] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 02/01/2023]
Abstract
Background: It has been proposed that GAS may form biofilms. Biofilms are microbial communities that aggregate on a surface, and exist within a self-produced matrix of extracellular polymeric substances. Biofilms offer bacteria an increased survival advantage, in which bacteria persist, and resist host immunity and antimicrobial treatment. The biofilm phenotype has long been recognized as a virulence mechanism for many Gram-positive and Gram-negative bacteria, however very little is known about the role of biofilms in GAS pathogenesis. Objective: This review provides an overview of the current knowledge of biofilms in GAS pathogenesis. This review assesses the evidence of GAS biofilm formation, the role of GAS virulence factors in GAS biofilm formation, modelling GAS biofilms, and discusses the polymicrobial nature of biofilms in the oropharynx in relation to GAS. Conclusion: Further study is needed to improve the current understanding of GAS as both a mono-species biofilm, and as a member of a polymicrobial biofilm. Improved modelling of GAS biofilm formation in settings closely mimicking in vivo conditions will ensure that biofilms generated in the lab closely reflect those occurring during clinical infection.
Collapse
Affiliation(s)
- Heema K N Vyas
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Emma-Jayne Proctor
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Jason McArthur
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Jody Gorman
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Martina Sanderson-Smith
- School of Chemistry and Molecular Bioscience, and Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| |
Collapse
|
8
|
Guo G, Wei D, Zhang Y, Wang K, Liu R, Wu Z, Kong L, Cheng L, Zhang W. M-like protein SrM is not crucial to the virulence of a novel isolate of Streptococcus equi subsp. ruminatorum from Macaca mulatta. Res Vet Sci 2020; 132:221-228. [PMID: 32615343 DOI: 10.1016/j.rvsc.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/27/2020] [Accepted: 05/04/2020] [Indexed: 11/28/2022]
Abstract
In this study, a Streptococcus strainnamed FJ1804, was isolated from a blood sample collected from a dead Macaca mulatta in China and, was subsequently classified as Streptococcus equi subsp. ruminatorum (S.e. ruminatorum) through 16S rRNA gene sequence analysis. After whole genome sequencing and analysis, an M-like protein encoding gene that encodes an SrM protein that is homologous to the crucial S.e. zooepidemicus crucial virulence factor SzP, was identified in the genome of FJ1804. To determinethe function of SrM in this bacterium, a strain deleted of srm as well as a complement strain were constructed. The results of in vitro cell adherence, invasion and phagocytosis assays and in vivo animal challenge and histopathology showed that the anti-phagocytosis was decreased and the adherence rate was increased in the srm deletion strain, whereas the invasion rate, pathological features and LD50 values inboth zebrafish and BALB/c mice model showed no difference compared to that observed for the WT strain. To the best of our knowledge, this is first of an infection caused by S.e. ruminatorum, which is a newly identified zoonotic pathogen, in Macaca mulatta, and our data suggest that, compared with other S.e. zooepidemicus strains, the SzP homologous protein is not crucial to the virulence of this bacterium.
Collapse
Affiliation(s)
- Genglin Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Dan Wei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Yuhang Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| | - Kaicheng Wang
- China Animal Health and Epidemiology Center, Qingdao, Shandong, China.
| | - Rongchang Liu
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian 350013, China.
| | - Zhiqiang Wu
- City Park Management Office of Nanping City, Nanping, Fujian 353000, China.
| | - Lifang Kong
- City Park Management Office of Nanping City, Nanping, Fujian 353000, China.
| | - Longfei Cheng
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian 350013, China.
| | - Wei Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing 210095, China; OIE Reference Lab for Swine Streptococcosis, Nanjing 210095, China.
| |
Collapse
|
9
|
Nye TM, Jacob KM, Holley EK, Nevarez JM, Dawid S, Simmons LA, Watson ME. DNA methylation from a Type I restriction modification system influences gene expression and virulence in Streptococcus pyogenes. PLoS Pathog 2019; 15:e1007841. [PMID: 31206562 PMCID: PMC6597129 DOI: 10.1371/journal.ppat.1007841] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/27/2019] [Accepted: 05/14/2019] [Indexed: 11/26/2022] Open
Abstract
DNA methylation is pervasive across all domains of life. In bacteria, the presence of N6-methyladenosine (m6A) has been detected among diverse species, yet the contribution of m6A to the regulation of gene expression is unclear in many organisms. Here we investigated the impact of DNA methylation on gene expression and virulence within the human pathogen Streptococcus pyogenes, or Group A Streptococcus. Single Molecule Real-Time sequencing and subsequent methylation analysis identified 412 putative m6A sites throughout the 1.8 Mb genome. Deletion of the Restriction, Specificity, and Methylation gene subunits (ΔRSM strain) of a putative Type I restriction modification system lost all detectable m6A at the recognition sites and failed to prevent transformation with foreign-methylated DNA. RNA-sequencing identified 20 genes out of 1,895 predicted coding regions with significantly different gene expression. All of the differentially expressed genes were down regulated in the ΔRSM strain relative to the parent strain. Importantly, we found that the presence of m6A DNA modifications affected expression of Mga, a master transcriptional regulator for multiple virulence genes, surface adhesins, and immune-evasion factors in S. pyogenes. Using a murine subcutaneous infection model, mice infected with the ΔRSM strain exhibited an enhanced host immune response with larger skin lesions and increased levels of pro-inflammatory cytokines compared to mice infected with the parent or complemented mutant strains, suggesting alterations in m6A methylation influence virulence. Further, we found that the ΔRSM strain showed poor survival within human neutrophils and reduced adherence to human epithelial cells. These results demonstrate that, in addition to restriction of foreign DNA, gram-positive bacteria also use restriction modification systems to regulate the expression of gene networks important for virulence. DNA methylation is common among many bacterial species, yet the contribution of DNA methylation to the regulation of gene expression is unclear outside of a limited number of gram-negative species. We characterized sites of DNA methylation throughout the genome of the gram-positive pathogen Streptococcus pyogenes or Group A Streptococcus. We determined that the gene products of a functional restriction modification system are responsible for genome-wide m6A. The mutant strain lacking DNA methylation showed altered gene expression compared to the parent strain, with several genes important for causing human disease down regulated. Furthermore, we showed that the mutant strain lacking DNA methylation exhibited altered virulence properties compared to the parent strain using various models of pathogenesis. The mutant strain was attenuated for both survival within human neutrophils and adherence to human epithelial cells, and was unable to suppress the host immune response in a murine subcutaneous infection model. Together, these results show that bacterial m6A contributes to differential gene expression and influences the ability of Group A Streptococcus to cause disease. DNA methylation is a conserved feature among bacteria and may represent a potential target for intervention in effort to interfere with the ability of bacteria to cause human disease.
Collapse
Affiliation(s)
- Taylor M. Nye
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Kristin M. Jacob
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Elena K. Holley
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Juan M. Nevarez
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Suzanne Dawid
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Lyle A. Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Michael E. Watson
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
10
|
The Second Messenger c-di-AMP Regulates Diverse Cellular Pathways Involved in Stress Response, Biofilm Formation, Cell Wall Homeostasis, SpeB Expression, and Virulence in Streptococcus pyogenes. Infect Immun 2019; 87:IAI.00147-19. [PMID: 30936159 DOI: 10.1128/iai.00147-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
Cyclic di-AMP (c-di-AMP) is a recently discovered second messenger in bacteria. The cellular level of c-di-AMP in Streptococcus pyogenes is predicted to be controlled by the synthase DacA and two putative phosphodiesterases, GdpP and Pde2. To investigate the role of c-di-AMP in S. pyogenes, we generated null mutants in each of these proteins by gene deletion. Unlike those in other Gram-positive pathogens such as Staphylococcus aureus and Listeria monocytogenes, DacA in S. pyogenes was not essential for growth in rich media. The DacA null mutant presented a growth defect that manifested through an increased lag time, produced no detectable biofilm, and displayed increased susceptibility toward environmental stressors such as high salt, low pH, reactive oxygen radicals, and cell wall-targeting antibiotics, suggesting that c-di-AMP plays significant roles in crucial cellular processes involved in stress management. The Pde2 null mutant exhibited a lower growth rate and increased biofilm formation, and interestingly, these phenotypes were distinct from those of the null mutant of GdpP, suggesting that Pde2 and GdpP play distinctive roles in c-di-AMP signaling. DacA and Pde2 were critical to the production of the virulence factor SpeB and to the overall virulence of S. pyogenes, as both DacA and Pde2 null mutants were highly attenuated in a mouse model of subcutaneous infection. Collectively, these results show that c-di-AMP is an important global regulator and is required for a proper response to stress and for virulence in S. pyogenes, suggesting that its signaling pathway could be an attractive antivirulence drug target against S. pyogenes infections.
Collapse
|
11
|
Nerlich A, Lapschies AM, Kohler TP, Cornax I, Eichhorn I, Goldmann O, Krienke P, Bergmann S, Nizet V, Hammerschmidt S, Rohde M, Fulde M. Homophilic protein interactions facilitate bacterial aggregation and IgG-dependent complex formation by the Streptococcus canis M protein SCM. Virulence 2019; 10:194-206. [PMID: 30829556 PMCID: PMC6527014 DOI: 10.1080/21505594.2019.1589362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Streptococcus canis is a zoonotic agent that causes serious invasive diseases in domestic animals and humans, but knowledge about its pathogenic potential and underlying virulence mechanisms is limited. Here, we report on the ability of certain S. canis isolates to form large bacterial aggregates when grown in liquid broth. Bacterial aggregation was attributed to the presence and the self-binding activity of SCM, the M protein of S. canis, as evaluated by bacterial sedimentation assays, immunofluorescence- and electron microscopic approaches. Using a variety of truncated recombinant SCM fragments, we demonstrated that homophilic SCM interactions occur via the N-terminal, but not the C-terminal part, of the mature M protein. Interestingly, when incubated in human plasma, SCM forms soluble protein complexes comprising its known ligands, immunoglobulin G (IgG) and plasminogen (Plg). Co-incubation studies with purified host proteins revealed that SCM-mediated complex formation is based on the interaction of SCM with itself and with IgG, but not with Plg or fibrinogen (Fbg), well-established constituents of M protein-mediated protein complexes in human-associated streptococci. Notably, these soluble, SCM-mediated plasma complexes harbored complement factor C1q, which can induce complement breakdown in the periphery and therefore represent another immune evasion mechanism of SCM.
Collapse
Affiliation(s)
- Andreas Nerlich
- a Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine , Charité Universitätsmedizin Berlin , Berlin , Germany.,b Department of Medical Microbiology , Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Antje-Maria Lapschies
- c Institute of Microbiology and Epizootics, Centre of Infection Medicine , Freie Universität Berlin , Berlin , Germany
| | - Thomas P Kohler
- d Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes , University of Greifswald , Greifswald , Germany
| | - Ingrid Cornax
- e Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences , UC San Diego , La Jolla , CA , USA
| | - Inga Eichhorn
- c Institute of Microbiology and Epizootics, Centre of Infection Medicine , Freie Universität Berlin , Berlin , Germany
| | - Oliver Goldmann
- f Infection Immunology Group, Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Petra Krienke
- c Institute of Microbiology and Epizootics, Centre of Infection Medicine , Freie Universität Berlin , Berlin , Germany
| | - Simone Bergmann
- b Department of Medical Microbiology , Helmholtz Centre for Infection Research , Braunschweig , Germany.,g Department of Infection Biology, Institute of Microbiology , Technische Universität Braunschweig , Braunschweig , Germany
| | - Victor Nizet
- e Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences , UC San Diego , La Jolla , CA , USA
| | - Sven Hammerschmidt
- d Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes , University of Greifswald , Greifswald , Germany
| | - Manfred Rohde
- b Department of Medical Microbiology , Helmholtz Centre for Infection Research , Braunschweig , Germany.,h Central Facility for Microscopy , Helmholtz Centre for Infection Research , Braunschweig , Germany
| | - Marcus Fulde
- b Department of Medical Microbiology , Helmholtz Centre for Infection Research , Braunschweig , Germany.,c Institute of Microbiology and Epizootics, Centre of Infection Medicine , Freie Universität Berlin , Berlin , Germany
| |
Collapse
|
12
|
Porayath C, Suresh MK, Biswas R, Nair BG, Mishra N, Pal S. Autolysin mediated adherence of Staphylococcus aureus with Fibronectin, Gelatin and Heparin. Int J Biol Macromol 2018; 110:179-184. [PMID: 29398086 PMCID: PMC5864509 DOI: 10.1016/j.ijbiomac.2018.01.047] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Abstract
Major autolysin (Atl) of Staphylococcus aureusis a cell surface associated peptidoglycan hydrolase with amidase and glucosaminidase domains. Atl enzymes (amidase and glucosaminidase) are known to participate in biofilm formation and also can bind with host matrices. Earlier studies demonstrated the binding of Atlwithfibronectin, thrombospondin 1, vitronectin and heat shock cognate protein Hsc70. Here, we have shown, Atl mediates attachment of S.aureus to heparin and gelatine as well. The atl mutant strain demonstrated around 2.5 fold decreased adherence with fibronectin, gelatin and heparin coated microtiter plates. The microscopic studies confirmed the reduced binding of atl mutant with them compared to its parental wild type and complemented mutant strains. Amidase and glucosaminidase were expressed as N-terminal histidine tagged proteins from Escherichia coli, purified and refolded. We found refolded amidase bind with fibronectin, gelatin and heparin; whereas refolded glucosaminidase binds with only fibronectin and heparin but not gelatin. These results reemphasize Atl as one of the crucial proteins from Staphylococcus that facilitate their binding with multiple host cellular components during colonization and infection.
Collapse
Affiliation(s)
- Chandni Porayath
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Maneesha K Suresh
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Cochin 682041, Kerala, India
| | - Raja Biswas
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Cochin 682041, Kerala, India
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Sanjay Pal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India.
| |
Collapse
|
13
|
Sjöholm K, Kilsgård O, Teleman J, Happonen L, Malmström L, Malmström J. Targeted Proteomics and Absolute Protein Quantification for the Construction of a Stoichiometric Host-Pathogen Surface Density Model. Mol Cell Proteomics 2017; 16:S29-S41. [PMID: 28183813 PMCID: PMC5393399 DOI: 10.1074/mcp.m116.063966] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/23/2017] [Indexed: 11/06/2022] Open
Abstract
Sepsis is a systemic immune response responsible for considerable morbidity and mortality. Molecular modeling of host-pathogen interactions in the disease state represents a promising strategy to define molecular events of importance for the transition from superficial to invasive infectious diseases. Here we used the Gram-positive bacterium Streptococcus pyogenes as a model system to establish a mass spectrometry based workflow for the construction of a stoichiometric surface density model between the S. pyogenes surface, the surface virulence factor M-protein, and adhered human blood plasma proteins. The workflow relies on stable isotope labeled reference peptides and selected reaction monitoring mass spectrometry analysis of a wild-type strain and an M-protein deficient mutant strain, to generate absolutely quantified protein stoichiometry ratios between S. pyogenes and interacting plasma proteins. The stoichiometry ratios in combination with a novel targeted mass spectrometry method to measure cell numbers enabled the construction of a stoichiometric surface density model using protein structures available from the protein data bank. The model outlines the topology and density of the host-pathogen protein interaction network on the S. pyogenes bacterial surface, revealing a dense and highly organized protein interaction network. Removal of the M-protein from S. pyogenes introduces a drastic change in the network topology, validated by electron microscopy. We propose that the stoichiometric surface density model of S. pyogenes in human blood plasma represents a scalable framework that can continuously be refined with the emergence of new results. Future integration of new results will improve the understanding of protein-protein interactions and their importance for bacterial virulence. Furthermore, we anticipate that the general properties of the developed workflow will facilitate the production of stoichiometric surface density models for other types of host-pathogen interactions.
Collapse
Affiliation(s)
- Kristoffer Sjöholm
- From the ‡Department of Immunotechnology, Faculty of Engineering, Lund University, Sweden
- §Division of Infection Medicine, Department of Clinical Sciences, Lund University, Sweden
| | - Ola Kilsgård
- §Division of Infection Medicine, Department of Clinical Sciences, Lund University, Sweden
| | - Johan Teleman
- §Division of Infection Medicine, Department of Clinical Sciences, Lund University, Sweden
| | - Lotta Happonen
- §Division of Infection Medicine, Department of Clinical Sciences, Lund University, Sweden
| | | | - Johan Malmström
- §Division of Infection Medicine, Department of Clinical Sciences, Lund University, Sweden;
| |
Collapse
|
14
|
Port GC, Cusumano ZT, Tumminello PR, Caparon MG. SpxA1 and SpxA2 Act Coordinately To Fine-Tune Stress Responses and Virulence in Streptococcus pyogenes. mBio 2017; 8:e00288-17. [PMID: 28351920 PMCID: PMC5371413 DOI: 10.1128/mbio.00288-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/01/2017] [Indexed: 11/23/2022] Open
Abstract
SpxA is a unique transcriptional regulator highly conserved among members of the phylum Firmicutes that binds RNA polymerase and can act as an antiactivator. Why some Firmicutes members have two highly similar SpxA paralogs is not understood. Here, we show that the SpxA paralogs of the pathogen Streptococcus pyogenes, SpxA1 and SpxA2, act coordinately to regulate virulence by fine-tuning toxin expression and stress resistance. Construction and analysis of mutants revealed that SpxA1- mutants were defective for growth under aerobic conditions, while SpxA2- mutants had severely attenuated responses to multiple stresses, including thermal and oxidative stresses. SpxA1- mutants had enhanced resistance to the cationic antimicrobial molecule polymyxin B, while SpxA2- mutants were more sensitive. In a murine model of soft tissue infection, a SpxA1- mutant was highly attenuated. In contrast, the highly stress-sensitive SpxA2- mutant was hypervirulent, exhibiting more extensive tissue damage and a greater bacterial burden than the wild-type strain. SpxA1- attenuation was associated with reduced expression of several toxins, including the SpeB cysteine protease. In contrast, SpxA2- hypervirulence correlated with toxin overexpression and could be suppressed to wild-type levels by deletion of speB These data show that SpxA1 and SpxA2 have opposing roles in virulence and stress resistance, suggesting that they act coordinately to fine-tune toxin expression in response to stress. SpxA2- hypervirulence also shows that stress resistance is not always essential for S. pyogenes pathogenesis in soft tissue.IMPORTANCE For many pathogens, it is generally assumed that stress resistance is essential for pathogenesis. For Streptococcus pyogenes, environmental stress is also used as a signal to alter toxin expression. The amount of stress likely informs the bacterium of the strength of the host's defense response, allowing it to adjust its toxin expression to produce the ideal amount of tissue damage, balancing between too little damage, which will result in its elimination, and too much damage, which will debilitate the host. Here we identify components of a genetic circuit involved in stress resistance and toxin expression that has a fine-tuning function in tissue damage. The circuit consists of two versions of the protein SpxA that regulate transcription and are highly similar but have opposing effects on the severity of soft tissue damage. These results will help us understand how virulence is fine-tuned in other pathogens that have two SpxA proteins.
Collapse
Affiliation(s)
- Gary C Port
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Zachary T Cusumano
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Paul R Tumminello
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Michael G Caparon
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
Global Analysis and Comparison of the Transcriptomes and Proteomes of Group A Streptococcus Biofilms. mSystems 2016; 1:mSystems00149-16. [PMID: 27933318 PMCID: PMC5141267 DOI: 10.1128/msystems.00149-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/01/2016] [Indexed: 11/20/2022] Open
Abstract
Prokaryotes are thought to regulate their proteomes largely at the level of transcription. However, the results from this first set of global transcriptomic and proteomic analyses of paired microbial samples presented here show that this assumption is false for the majority of genes and their products in S. pyogenes. In addition, the tenuousness of the link between transcription and translation becomes even more pronounced when microbes exist in a biofilm or a stationary planktonic state. Since the transcriptome level does not usually equal the proteome level, the validity attributed to gene expression studies as well as proteomic studies in microbial analyses must be brought into question. Therefore, the results attained by either approach, whether RNA-seq or shotgun proteomics, must be taken in context and evaluated with particular care since they are by no means interchangeable. To gain a better understanding of the genes and proteins involved in group A Streptococcus (GAS; Streptococcus pyogenes) biofilm growth, we analyzed the transcriptome, cellular proteome, and cell wall proteome from biofilms at different stages and compared them to those of plankton-stage GAS. Using high-throughput RNA sequencing (RNA-seq) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) shotgun proteomics, we found distinct expression profiles in the transcriptome and proteome. A total of 46 genes and 41 proteins showed expression across the majority of biofilm time points that was consistently higher or consistently lower than that seen across the majority of planktonic time points. However, there was little overlap between the genes and proteins on these two lists. In line with other studies comparing transcriptomic and proteomic data, the overall correlation between the two data sets was modest. Furthermore, correlation was poorest for biofilm samples. This suggests a high degree of regulation of protein expression by nontranscriptional mechanisms. This report illustrates the benefits and weaknesses of two different approaches to global expression profiling, and it also demonstrates the advantage of using proteomics in conjunction with transcriptomics to gain a more complete picture of global expression within biofilms. In addition, this report provides the fullest characterization of expression patterns in GAS biofilms currently available. IMPORTANCE Prokaryotes are thought to regulate their proteomes largely at the level of transcription. However, the results from this first set of global transcriptomic and proteomic analyses of paired microbial samples presented here show that this assumption is false for the majority of genes and their products in S. pyogenes. In addition, the tenuousness of the link between transcription and translation becomes even more pronounced when microbes exist in a biofilm or a stationary planktonic state. Since the transcriptome level does not usually equal the proteome level, the validity attributed to gene expression studies as well as proteomic studies in microbial analyses must be brought into question. Therefore, the results attained by either approach, whether RNA-seq or shotgun proteomics, must be taken in context and evaluated with particular care since they are by no means interchangeable.
Collapse
|
16
|
Brouwer S, Barnett TC, Rivera-Hernandez T, Rohde M, Walker MJ. Streptococcus pyogenes adhesion and colonization. FEBS Lett 2016; 590:3739-3757. [PMID: 27312939 DOI: 10.1002/1873-3468.12254] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 12/19/2022]
Abstract
Streptococcus pyogenes (group A Streptococcus, GAS) is a human-adapted pathogen responsible for a wide spectrum of disease. GAS can cause relatively mild illnesses, such as strep throat or impetigo, and less frequent but severe life-threatening diseases such as necrotizing fasciitis and streptococcal toxic shock syndrome. GAS is an important public health problem causing significant morbidity and mortality worldwide. The main route of GAS transmission between humans is through close or direct physical contact, and particularly via respiratory droplets. The upper respiratory tract and skin are major reservoirs for GAS infections. The ability of GAS to establish an infection in the new host at these anatomical sites primarily results from two distinct physiological processes, namely bacterial adhesion and colonization. These fundamental aspects of pathogenesis rely upon a variety of GAS virulence factors, which are usually under strict transcriptional regulation. Considerable progress has been made in better understanding these initial infection steps. This review summarizes our current knowledge of the molecular mechanisms of GAS adhesion and colonization.
Collapse
Affiliation(s)
- Stephan Brouwer
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Timothy C Barnett
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre For Infection Research, Braunschweig, Germany
| | - Mark J Walker
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| |
Collapse
|
17
|
Hendrickson C, Euler CW, Nguyen SV, Rahman M, McCullor KA, King CJ, Fischetti VA, McShan WM. Elimination of Chromosomal Island SpyCIM1 from Streptococcus pyogenes Strain SF370 Reverses the Mutator Phenotype and Alters Global Transcription. PLoS One 2015; 10:e0145884. [PMID: 26701803 PMCID: PMC4689407 DOI: 10.1371/journal.pone.0145884] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/09/2015] [Indexed: 02/04/2023] Open
Abstract
Streptococcus pyogenes chromosomal island M1 (SpyCIM1) integrates by site-specific recombination into the 5’ end of DNA mismatch repair (MMR) gene mutL in strain SF370SmR, blocking transcription of it and the downstream operon genes. During exponential growth, SpyCIM1 excises from the chromosome and replicates as an episome, restoring mutL transcription. This process is reversed in stationary phase with SpyCIM1 re-integrating into mutL, returning the cells to a mutator phenotype. Here we show that elimination of SpyCIM1 relieves this mutator phenotype. The downstream MMR operon genes, multidrug efflux pump lmrP, Holliday junction resolution helicase ruvA, and DNA base excision repair glycosylase tag, are also restored to constitutive expression by elimination of SpyCIM1. The presence of SpyCIM1 alters global transcription patterns in SF370SmR. RNA sequencing (RNA-Seq) demonstrated that loss of SpyCIM1 in the SpyCIM1 deletion mutant, CEM1Δ4, impacted the expression of over 100 genes involved in virulence and metabolism both in early exponential phase, when the SpyCIM1 is episomal, as well as at the onset of stationary phase, when SpyCIM1 has reintegrated into mutL. Among these changes, the up-regulation of the genes for the antiphagocytic M protein (emm1), streptolysin O (slo), capsule operon (hasABC), and streptococcal pyrogenic exotoxin (speB), are particularly notable. The expression pattern of the MMR operon confirmed our earlier observations that these genes are transcribed in early exponential phase but silenced as stationary phase is approached. Thus, the direct role of SpyCIM1 in causing the mutator phenotype is confirmed, and further, its influence upon the biology of S. pyogenes was found to impact multiple genes in addition to the MMR operon, which is a novel function for a mobile genetic element. We suggest that such chromosomal islands are a remarkable evolutionary adaptation to promote the survival of its S. pyogenes host cell in changing environments.
Collapse
Affiliation(s)
- Christina Hendrickson
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- The Biology Department, The University of Central Oklahoma, Edmond, Oklahoma, United States of America
| | - Chad W. Euler
- Laboratory of Bacterial Pathogenesis and Immunology, The Rockefeller University, New York, New York, United States of America
- Department of Medical Laboratory Sciences, Belfer Research Building, Hunter College, CUNY, New York, New York, United States of America
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, United States of America
| | - Scott V. Nguyen
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Maliha Rahman
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Kimberly A. McCullor
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Catherine J. King
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Vincent A. Fischetti
- Laboratory of Bacterial Pathogenesis and Immunology, The Rockefeller University, New York, New York, United States of America
| | - W. Michael McShan
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
18
|
Rapid Lymphatic Dissemination of Encapsulated Group A Streptococci via Lymphatic Vessel Endothelial Receptor-1 Interaction. PLoS Pathog 2015; 11:e1005137. [PMID: 26352587 PMCID: PMC4564194 DOI: 10.1371/journal.ppat.1005137] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/10/2015] [Indexed: 11/29/2022] Open
Abstract
The host lymphatic network represents an important conduit for pathogen dissemination. Indeed, the lethal human pathogen group A streptococcus has a predilection to induce pathology in the lymphatic system and draining lymph nodes, however the underlying basis and subsequent consequences for disease outcome are currently unknown. Here we report that the hyaluronan capsule of group A streptococci is a crucial virulence determinant for lymphatic tropism in vivo, and further, we identify the lymphatic vessel endothelial receptor-1 as the critical host receptor for capsular hyaluronan in the lymphatic system. Interference with this interaction in vivo impeded bacterial dissemination to local draining lymph nodes and, in the case of a hyper-encapsulated M18 strain, redirected streptococcal entry into the blood circulation, suggesting a pivotal role in the manifestation of streptococcal infections. Our results reveal a novel function for bacterial capsular polysaccharide in directing lymphatic tropism, with potential implications for disease pathology. Pathogens are known to invade the host not only via the systemic circulation but also via the lymphatic network, however the mechanisms underlying the latter route and the consequences for disease outcome have not been well studied. The important human pathogen, group A streptococcus, is responsible for a number of clinical syndromes affecting both the lymphatic vessels and draining lymph nodes, such as lymphangitis and lymphadenitis. How such pathologies are orchestrated, and their significance in the development of serious infection are currently unknown. In this study, we show that the hyaluronan capsule secreted by group A streptococcus is critical for bacterial spread to draining lymph nodes, and we demonstrate that this occurs as a result of a specific interaction with the lymphatic vessel endothelial receptor-1. Genetic deletion or functional blockade of this receptor prevented streptococcal transit to draining lymph nodes in a murine model of infection, which in turn enhanced bacterial spread into the blood circulation. Together these results define a novel interaction between the group A streptococcal capsule and the lymphatic endothelial receptor-1 as a critical axis in the establishment of lymphatic tropism for this pathogen, with clear implications for disease severity in the host.
Collapse
|
19
|
Inhibition of microbial adhesion to plastic surface and human buccal epithelial cells by Rhodomyrtus tomentosa leaf extract. Arch Oral Biol 2014; 59:1256-65. [PMID: 25146902 DOI: 10.1016/j.archoralbio.2014.07.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 05/02/2014] [Accepted: 07/22/2014] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The adherence of oral pathogenic microorganisms to host tissues is the initial step for successful process of oral diseases. This study aimed to determine the effect of the Rhodomyrtus tomentosa leaf extract and rhodomyrtone, an antibacterial compound from R. tomentosa leaf, on adhesion of some oral pathogens to polystyrene plastic surface and human buccal epithelial cells. METHODS The minimum inhibitory concentration (MIC) was evaluated using broth microdilution method. The microbial adhesion to the plastic surface and buccal cells was determined using microtiter plate method and microscopy technique. RESULTS The ethanol extract of leaf demonstrated antibacterial activity against oral microorganisms including Staphylococcus aureus ATCC 25923, Streptococcus mutans (clinical isolate), and Candida albicans ATCC 90028 with the MIC values of 31.25, 15.62, and 1000μg/ml, respectively. Rhodomyrtone displayed activity with the MIC values of 0.78 and 0.39μg/ml against S. aureus ATCC 25923 and S. mutans, respectively. The MIC value of the compound against C. albicans ATCC 90028 was more than 100μg/ml which was the highest test concentration. All pathogenic microorganisms treated with the extract and rhodomyrtone at their subinhibitory concentrations resulted in a decrease in their adherence ability to both plastic surface and buccal cells. CONCLUSION It is suggested that R. tomentosa extract and rhodomyrtone may be useful in therapy or as prophylaxis in infections involving oral pathogens.
Collapse
|
20
|
Streptococcus pyogenes polymyxin B-resistant mutants display enhanced ExPortal integrity. J Bacteriol 2014; 196:2563-77. [PMID: 24794568 DOI: 10.1128/jb.01596-14] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The ExPortal protein secretion organelle in Streptococcus pyogenes is an anionic phospholipid-containing membrane microdomain enriched in Sec translocons and postsecretion protein biogenesis factors. Polymyxin B binds to and disrupts ExPortal integrity, resulting in defective secretion of several toxins. To gain insight into factors that influence ExPortal organization, a genetic screen was conducted to select for spontaneous polymyxin B-resistant mutants displaying enhanced ExPortal integrity. Whole-genome resequencing of 25 resistant mutants revealed from one to four mutations per mutant genome clustered primarily within a core set of 10 gene groups. Construction of mutants with individual deletions or insertions demonstrated that 7 core genes confer resistance and enhanced ExPortal integrity through loss of function, while 3 were likely due to gain of function and/or combinatorial effects. Core resistance genes include a transcriptional regulator of lipid biosynthesis, several genes involved in nutrient acquisition, and a variety of genes involved in stress responses. Two members of the latter class also function as novel regulators of the secreted SpeB cysteine protease. Analysis of the most frequently isolated mutation, a single nucleotide deletion in a track of 9 consecutive adenine residues in pstS, encoding a component of a high-affinity Pi transporter, suggests that this sequence functions as a molecular switch to facilitate stress adaptation. Together, these data suggest the existence of a membrane stress response that promotes enhanced ExPortal integrity and resistance to cationic antimicrobial peptides.
Collapse
|
21
|
Sjöholm K, Karlsson C, Linder A, Malmström J. A comprehensive analysis of the Streptococcus pyogenes and human plasma protein interaction network. MOLECULAR BIOSYSTEMS 2014; 10:1698-708. [PMID: 24525632 DOI: 10.1039/c3mb70555b] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Streptococcus pyogenes is a major human bacterial pathogen responsible for severe and invasive disease associated with high mortality rates. The bacterium interacts with several human blood plasma proteins and clarifying these interactions and their biological consequences will help to explain the progression from mild to severe infections. In this study, we used a combination of mass spectrometry (MS) based techniques to comprehensively quantify the components of the S. pyogenes-plasma protein interaction network. From an initial list of 181 interacting human plasma proteins defined using liquid chromatography (LC)-MS/MS analysis we further subdivided the interacting protein list using selected reaction monitoring (SRM) depending on the level of enrichment and protein concentration on the bacterial surface. The combination of MS methods revealed several previously characterized interactions between the S. pyogenes surface and human plasma along with many more, so far uncharacterised, possible plasma protein interactions with S. pyogenes. In follow-up experiments, the combination of MS techniques was applied to study differences in protein binding to a S. pyogenes wild type strain and an isogenic mutant lacking several important virulence factors, and a unique pair of invasive and non-invasive S. pyogenes isolates from the same patient. Comparing the plasma protein-binding properties of the wild type and the mutant and the invasive and non-invasive S. pyogenes bacteria revealed considerable differences, underlining the significance of these protein interactions. The results also demonstrate the power of the developed mass spectrometry method to investigate host-microbial relationships with a large proteomics depth and high quantitative accuracy.
Collapse
Affiliation(s)
- Kristoffer Sjöholm
- Department of Immunotechnology, Faculty of Engineering, Lund University, Sweden
| | | | | | | |
Collapse
|
22
|
Georgousakis MM, McMillan DJ, Batzloff MR, Sriprakash KS. Moving forward: a mucosal vaccine against group A streptococcus. Expert Rev Vaccines 2014; 8:747-60. [DOI: 10.1586/erv.09.33] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
23
|
An association between peptidoglycan synthesis and organization of the Streptococcus pyogenes ExPortal. mBio 2013; 4:e00485-13. [PMID: 24065630 PMCID: PMC3781834 DOI: 10.1128/mbio.00485-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ExPortal of Streptococcus pyogenes is a focal microdomain of the cytoplasmic membrane that clusters the translocons of the general secretory pathway with accessory factors to facilitate the maturation of secreted polypeptides. While it is known that the ExPortal is enriched in anionic lipids, the mechanisms that organize the ExPortal are poorly understood. In the present study, we examined the role of the cell wall in organizing and maintaining the ExPortal. Removal of the cell wall resulted in a loss of ExPortal focal integrity accompanied by the circumferential redistribution of ExPortal lipid and protein components. A similar loss occurred upon treatment with gallidermin, a nonpermeabilizing lantibiotic that targets the lipid II precursor of peptidoglycan synthesis, and this treatment disrupted the secretion of several ExPortal substrates. Furthermore, several enzymes involved in the membrane-associated steps of lipid II synthesis, including MraY and MurN, were found to localize to a single discrete focus in the membrane that was coincident with the focal location of the secretory translocons and the anionic lipid microdomain. These data suggest that the ExPortal is associated with the site of peptidoglycan precursor synthesis and that peptidoglycan biogenesis influences ExPortal organization. These data add to an emerging literature indicating that cell wall biogenesis, cell division, and protein secretion are spatially coorganized processes. Since Gram-positive bacteria lack a periplasmic space, they lack a protected compartment to spatially coordinate interaction between newly secreted proteins and the factors required to process them. This represents a significant problem for pathogens that depend on the secretion of toxins and cell wall-associated adhesins to cause disease. Streptococci solve this dilemma by restricting secretion and processing factors to a defined region of the membrane. However, the mechanisms that promote restriction are not understood. In this study, we show that restriction of these factors in the pathogen Streptococcus pyogenes is intimately linked with the presence of the cell wall and its synthesis. Furthermore, several cell wall synthesis proteins are also restricted to the site of protein secretion. This study contributes to our understanding of how the Gram-positive cell is organized to coordinate protein secretion and biogenesis with cell wall synthesis and to the ongoing development of antibiotics that target these processes.
Collapse
|
24
|
Catalanotti P, Catania MR, Lucido M, Martini S, Gallè F, Ortega De Luna L, Rossano F. T Serotyping and Genomic Profile of Erythromycin- Resistant or -SensitiveStreptococcus pyogenesIsolated in Campania Region, Italy. J Chemother 2013; 17:131-7. [PMID: 15920897 DOI: 10.1179/joc.2005.17.2.131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Streptococcus pyogenes causes mild infections, such as pharyngitis, and severe infections, such as necrotizing fascitis. In recent years, erythromycin-resistant strains of S. pyogenes have been reported in many countries. In some areas of Italy, increased rates of erythromycin resistance were first observed in the mid-1990s. Here, we report epidemiological T serotyping, invasiveness, erythromycin resistance, and PFGE patterns of 99 S. pyogenes strains isolated at the Laboratory of Clinical Microbiology of the Second University of Naples, Italy. Regarding T serotyping, 26 of 99 strains were W+, 16 strains were U+, 16 were X+, and 14 were agglutinated by anti T serum. A low percentage revealed Y+. Twelve strains were not T serotyped. PFGE patterns showed species polymorphism; however, inside the various serotypes, we demonstrated a fair homogeneity. No correlation among invasiveness and T serotype or PFGE pattern has been shown. Twenty-two of 99 strains were erythromycin-resistant.
Collapse
Affiliation(s)
- P Catalanotti
- Dipartimento di Medicina Sperimentale, Sezione di Microbiologia e Microbiologia Clinica, Seconda Università di Napoli, Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
25
|
Watson ME, Nielsen HV, Hultgren SJ, Caparon MG. Murine vaginal colonization model for investigating asymptomatic mucosal carriage of Streptococcus pyogenes. Infect Immun 2013; 81:1606-17. [PMID: 23460515 PMCID: PMC3648019 DOI: 10.1128/iai.00021-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/21/2013] [Indexed: 11/20/2022] Open
Abstract
While many virulence factors promoting Streptococcus pyogenes invasive disease have been described, specific streptococcal factors and host properties influencing asymptomatic mucosal carriage remain uncertain. To address the need for a refined model of prolonged S. pyogenes asymptomatic mucosal colonization, we have adapted a preestrogenized murine vaginal colonization model for S. pyogenes. In this model, derivatives of strains HSC5, SF370, JRS4, NZ131, and MEW123 established a reproducible, asymptomatic colonization of the vaginal mucosa over a period of typically 3 to 4 weeks' duration at a relatively high colonization efficiency. Prior treatment with estradiol prolonged streptococcal colonization and was associated with reduced inflammation in the colonized vaginal epithelium as well as a decreased leukocyte presence in vaginal fluid compared to the levels of inflammation and leukocyte presence in non-estradiol-treated control mice. The utility of our model for investigating S. pyogenes factors contributing to mucosal carriage was verified, as a mutant with a mutation in the transcriptional regulator catabolite control protein A (CcpA) demonstrated significant impairment in vaginal colonization. An assessment of in vivo transcriptional activity in the CcpA(-) strain for several known CcpA-regulated genes identified significantly elevated transcription of lactate oxidase (lctO) correlating with excessive generation of hydrogen peroxide to self-lethal levels. Deletion of lctO did not impair colonization, but deletion of lctO in a CcpA(-) strain prolonged carriage, exceeding even that of the wild-type strain. Thus, while LctO is not essential for vaginal colonization, its dysregulation is deleterious, highlighting the critical role of CcpA in promoting mucosal colonization. The vaginal colonization model should prove effective for future analyses of S. pyogenes mucosal colonization.
Collapse
Affiliation(s)
- Michael E. Watson
- Divison of Pediatric Infectious Diseases, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hailyn V. Nielsen
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, USA
| | - Scott J. Hultgren
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael G. Caparon
- Department of Molecular Microbiology, Washington University Medical School, St. Louis, Missouri, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Rohde M, Chhatwal GS. Adherence and invasion of streptococci to eukaryotic cells and their role in disease pathogenesis. Curr Top Microbiol Immunol 2012. [PMID: 23203001 DOI: 10.1007/82_2012_281] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Streptococcal adhesion, invasion, intracellular trafficking, dissemination, and persistence in eukaryotic cells have a variety of implications in the infection pathogenesis. While cell adhesion establishes the initial host contact, adhering bacteria exploit the host cell for their own benefit. Internalization into the host cell is an essential step for bacterial survival and subsequent dissemination and persistence, thus playing a key role in the course of infection. This chapter summarizes the current knowledge about the diverse mechanisms of streptococcal adhesion to and invasion into different eukaryotic cells and the impact on dissemination and persistence which is reflected by consequences for the pathogenesis of streptococcal infections.
Collapse
Affiliation(s)
- Manfred Rohde
- Department of Medical Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | | |
Collapse
|
27
|
Limsuwan S, Hesseling-Meinders A, Voravuthikunchai SP, van Dijl JM, Kayser O. Potential antibiotic and anti-infective effects of rhodomyrtone from Rhodomyrtus tomentosa (Aiton) Hassk. on Streptococcus pyogenes as revealed by proteomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:934-940. [PMID: 21439802 DOI: 10.1016/j.phymed.2011.02.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 12/28/2010] [Accepted: 02/07/2011] [Indexed: 05/30/2023]
Abstract
Rhodomyrtone from Rhodomyrtus tomentosa (Aiton) Hassk. leaf extract has a strong antibacterial activity against the bacterial pathogen Streptococcus pyogenes. Our previous studies indicated that the bactericidal activity of rhodomyrtone might involve intracellular targets. In the present studies we followed a proteomics approach to investigate the mode of action of rhodomyrtone on S. pyogenes. For this purpose, S. pyogenes was cultivated in the presence of 0.39 μg/ml rhodomyrtone, which corresponds to 50% of the minimal inhibitory concentration. The results show that the amounts of various enzymes associated with important metabolic pathways were strongly affected, which is consistent with the growth-inhibiting effect of rhodomyrtone. Additionally, cells of S. pyogenes grown in the presence of rhodomyrtone produced reduced amounts of known virulence factors, such as the glyceraldehyde-3-phosphate dehydrogenase, the CAMP factor, and the streptococcal pyrogenic exotoxin C. Taken together, these findings indicate that rhodomyrtone has both antimicrobial and anti-infective activities, which make it an interesting candidate drug.
Collapse
Affiliation(s)
- Surasak Limsuwan
- Faculty of Traditional Thai Medicine and Natural Products Research Center, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | | | | | | | | |
Collapse
|
28
|
Ghosh P. The nonideal coiled coil of M protein and its multifarious functions in pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 715:197-211. [PMID: 21557065 DOI: 10.1007/978-94-007-0940-9_12] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The M protein is a major virulence factor of Streptococcus pyogenes (group A Streptococcus, GAS). This gram-positive bacterial pathogen is responsible for mild infections, such as pharyngitis, and severe invasive disease, like streptococcal toxic shock syndrome. M protein contributes to GAS virulence in multifarious ways, including blocking deposition of antibodies and complement, helping formation of microcolonies, neutralizing antimicrobial peptides, and triggering a proinflammatory and procoagulatory state. These functions are specified by interactions between M protein and many host components, especially C4BP and fibrinogen. The former interaction is conserved among many antigenically variant M protein types but occurs in a strikingly sequence-independent manner, and the latter is associated in the M1 protein type with severe invasive disease. Remarkably for a protein of such diverse interactions, the M protein has a relatively simple but nonideal α-helical coiled coil sequence. This sequence nonideality is a crucial feature of M protein. Nonideal residues give rise to specific irregularities in its coiled-coil structure, which are essential for interactions with fibrinogen and establishment of a proinflammatory state. In addition, these structural irregularities are reminiscent of those in myosin and tropomyosin, which are targets for crossreactive antibodies in patients suffering from autoimmune sequelae of GAS infection.
Collapse
Affiliation(s)
- Partho Ghosh
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, 92093-0375, USA.
| |
Collapse
|
29
|
Lin A, Loughman JA, Zinselmeyer BH, Miller MJ, Caparon MG. Streptolysin S inhibits neutrophil recruitment during the early stages of Streptococcus pyogenes infection. Infect Immun 2009; 77:5190-201. [PMID: 19687200 PMCID: PMC2772533 DOI: 10.1128/iai.00420-09] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 05/24/2009] [Accepted: 08/10/2009] [Indexed: 11/20/2022] Open
Abstract
In contrast to infection of superficial tissues, Streptococcus pyogenes infection of deeper tissue can be associated with a significantly diminished inflammatory response, suggesting that this bacterium has the ability to both promote and suppress inflammation. To examine this, we analyzed the behavior of an S. pyogenes mutant deficient in expression of the cytolytic toxin streptolysin S (SLS-) and evaluated events that occur during the first few hours of infection by using several models including injection of zebrafish (adults, larvae, and embryos), a transepithelial polymorphonuclear leukocyte (PMN) migration assay, and two-photon microscopy of mice in vivo. In contrast to wild-type S. pyogenes, the SLS- mutant was associated with the robust recruitment of neutrophils and significantly reduced lethal myositis in adult zebrafish. Similarly, the mutant was attenuated in embryos in its ability to cause lethality. Infection of larva muscle allowed an analysis of inflammation in real time, which revealed that the mutant had recruited PMNs to the infection site. Analysis of transepithelial migration in vitro suggested that SLS inhibited the host cells' production of signals chemotactic for neutrophils, which contrasted with the proinflammatory effect of an unrelated cytolytic toxin, streptolysin O. Using two-photon microscopy of mice in vivo, we showed that the extravasation of neutrophils during infection with SLS- mutant bacteria was significantly accelerated compared to infection with wild-type S. pyogenes. Taken together, these data support a role for SLS in the inhibition of neutrophil recruitment during the early stages of S. pyogenes infection.
Collapse
Affiliation(s)
- Ada Lin
- Department of Pediatrics, Department of Pathology and Immunology, Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110-1093
| | - Jennifer A. Loughman
- Department of Pediatrics, Department of Pathology and Immunology, Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110-1093
| | - Bernd H. Zinselmeyer
- Department of Pediatrics, Department of Pathology and Immunology, Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110-1093
| | - Mark J. Miller
- Department of Pediatrics, Department of Pathology and Immunology, Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110-1093
| | - Michael G. Caparon
- Department of Pediatrics, Department of Pathology and Immunology, Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri 63110-1093
| |
Collapse
|
30
|
Araújo ÂMM, Oliveira ICMD, Mattos MCD, Benchetrit LC. Cell surface hydrophobicity and adherence of a strain of group B streptococci during the post-antibiotic effect of penicillin. Rev Inst Med Trop Sao Paulo 2008; 50:203-7. [DOI: 10.1590/s0036-46652008000400003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 06/25/2008] [Indexed: 11/22/2022] Open
Abstract
The minimum inhibitory concentration and post-antibiotic effects of an antimicrobial agent are parameters to be taken into consideration when determining its dosage schedules. The in vitro post-antibiotic effects on cell surface hydrophobicity and bacterial adherence were examined in one strain of group B streptococci. Exposure of the microorganism for 2 h at 37 °C to 1 x MIC of penicillin induced a PAE of 1.1 h. The cell surface charge of the Streptococcus was altered significantly during the post-antibiotic phase as shown by its ability to bind to xylene: hydrophobicity was decreased. Bacterial adherence to human buccal epithelial cells was also reduced. The results of the present investigation indicate that studies designed to determine therapeutic regimens should evaluate the clinical significance of aspects of bacterial physiology during the post-antibiotic period.
Collapse
|
31
|
Locke JB, Aziz RK, Vicknair MR, Nizet V, Buchanan JT. Streptococcus iniae M-like protein contributes to virulence in fish and is a target for live attenuated vaccine development. PLoS One 2008; 3:e2824. [PMID: 18665241 PMCID: PMC2483786 DOI: 10.1371/journal.pone.0002824] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Accepted: 06/17/2008] [Indexed: 12/21/2022] Open
Abstract
Background Streptococcus iniae is a significant pathogen in finfish aquaculture, though knowledge of virulence determinants is lacking. Through pyrosequencing of the S. iniae genome we have identified two gene homologues to classical surface-anchored streptococcal virulence factors: M-like protein (simA) and C5a peptidase (scpI). Methodology/Principal Findings S. iniae possesses a Mga-like locus containing simA and a divergently transcribed putative mga-like regulatory gene, mgx. In contrast to the Mga locus of group A Streptococcus (GAS, S. pyogenes), scpI is located distally in the chromosome. Comparative sequence analysis of the Mgx locus revealed only one significant variant, a strain with an insertion frameshift mutation in simA and a deletion mutation in a region downstream of mgx, generating an ORF which may encode a second putative mga-like gene, mgx2. Allelic exchange mutagenesis of simA and scpI was employed to investigate the potential role of these genes in S. iniae virulence. Our hybrid striped bass (HSB) and zebrafish models of infection revealed that M-like protein contributes significantly to S. iniae pathogenesis whereas C5a peptidase-like protein does not. Further, in vitro cell-based analyses indicate that SiMA, like other M family proteins, contributes to cellular adherence and invasion and provides resistance to phagocytic killing. Attenuation in our virulence models was also observed in the S. iniae isolate possessing a natural simA mutation. Vaccination of HSB with the ΔsimA mutant provided 100% protection against subsequent challenge with a lethal dose of wild-type (WT) S. iniae after 1,400 degree days, and shows promise as a target for live attenuated vaccine development. Conclusions/Significance Analysis of M-like protein and C5a peptidase through allelic replacement revealed that M-like protein plays a significant role in S. iniae virulence, and the Mga-like locus, which may regulate expression of this gene, has an unusual arrangement. The M-like protein mutant created in this research holds promise as live-attenuated vaccine.
Collapse
Affiliation(s)
- Jeffrey B. Locke
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Center for Marine Biotechnology & Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, United States of America
| | - Ramy K. Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mike R. Vicknair
- Kent SeaTech Corporation, San Diego, California, United States of America
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Center for Marine Biotechnology & Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, United States of America
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
| | - John T. Buchanan
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
- Aqua Bounty Technologies, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Gratz N, Siller M, Schaljo B, Pirzada ZA, Gattermeier I, Vojtek I, Kirschning CJ, Wagner H, Akira S, Charpentier E, Kovarik P. Group A streptococcus activates type I interferon production and MyD88-dependent signaling without involvement of TLR2, TLR4, and TLR9. J Biol Chem 2008; 283:19879-87. [PMID: 18480050 PMCID: PMC2459277 DOI: 10.1074/jbc.m802848200] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Indexed: 01/07/2023] Open
Abstract
Bacterial pathogens are recognized by the innate immune system through pattern recognition receptors, such as Toll-like receptors (TLRs). Engagement of TLRs triggers signaling cascades that launch innate immune responses. Activation of MAPKs and NF-kappaB, elements of the major signaling pathways induced by TLRs, depends in most cases on the adaptor molecule MyD88. In addition, Gram-negative or intracellular bacteria elicit MyD88-independent signaling that results in production of type I interferon (IFN). Here we show that in mouse macrophages, the activation of MyD88-dependent signaling by the extracellular Gram-positive human pathogen group A streptococcus (GAS; Streptococcus pyogenes) does not require TLR2, a receptor implicated in sensing of Gram-positive bacteria, or TLR4 and TLR9. Redundant engagement of either of these TLR molecules was excluded by using TLR2/4/9 triple-deficient macrophages. We further demonstrate that infection of macrophages by GAS causes IRF3 (interferon-regulatory factor 3)-dependent, MyD88-independent production of IFN. Surprisingly, IFN is induced also by GAS lacking slo and sagA, the genes encoding cytolysins that were shown to be required for IFN production in response to other Gram-positive bacteria. Our data indicate that (i) GAS is recognized by a MyD88-dependent receptor other than any of those typically used by bacteria, and (ii) GAS as well as GAS mutants lacking cytolysin genes induce type I IFN production by similar mechanisms as bacteria requiring cytoplasmic escape and the function of cytolysins.
Collapse
Affiliation(s)
- Nina Gratz
- Max F Perutz Laboratories, Department of Microbiology and Immunobiology, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
The signal recognition particle pathway is required for virulence in Streptococcus pyogenes. Infect Immun 2008; 76:2612-9. [PMID: 18411293 DOI: 10.1128/iai.00239-07] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The signal recognition particle (SRP) pathway is a universally conserved pathway for targeting polypeptides for secretion via the cotranslational pathway. In particular, the SRP pathway is thought to be the main mechanism for targeting polypeptides in gram-positive bacteria, including a number of important human pathogens. Though widely considered to be an essential cellular component, recent advances have indicated this pathway may be dispensable in gram-positive bacteria of the genus Streptococcus under in vitro conditions. However, its importance for the pathogenesis of streptococcal disease is unknown. In this study, we investigated the importance of the SRP pathway for virulence factor secretion in the human pathogen Streptococcus pyogenes. While the SRP pathway was not found to be essential for viability in vitro, SRP mutants demonstrated a medium-specific growth defect that could be rescued by the addition of glucose. We also observed that a distinct subset of virulence factors were dependent upon the SRP pathway for secretion, whereas others were completely independent of this pathway. Significantly, deletion of the SRP pathway resulted in mutants that were highly attenuated in both a zebrafish model of necrotic myositis and a murine subcutaneous ulcer model, highlighting the importance of this pathway in vivo. These studies emphasize the importance of the SRP pathway for the in vivo survival and pathogenesis of S. pyogenes.
Collapse
|
34
|
Abbot EL, Smith WD, Siou GPS, Chiriboga C, Smith RJ, Wilson JA, Hirst BH, Kehoe MA. Pili mediate specific adhesion of Streptococcus pyogenes to human tonsil and skin. Cell Microbiol 2007; 9:1822-33. [PMID: 17359232 DOI: 10.1111/j.1462-5822.2007.00918.x] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Very little is known about the biological functions of pili that have recently been found to be expressed by important Gram-positive pathogens such as Corynebacterium diphtheriae, Streptococcus agalacticae, S. pneumoniae and S. pyogenes. Using various ex vivo tissue and cellular models, here we show that pili mediate adhesion of serotype M1 S. pyogenes strain SF370 to both human tonsil epithelium and primary human keratinocytes, which represent the two main sites of infection by this human-specific pathogen. Mutants lacking minor pilus subunits retained the ability to express cell-surface pili, but these were functionally defective. In contrast to above, pili were not required for S. pyogenes adhesion to either immortalized HEp-2 or A549 cells, highlighting an important limitation of these extensively used adhesion/invasion models. Adhering bacteria were internalized very effectively by both HEp-2 and A549 cells, but not by tonsil epithelium or primary keratinocytes. While pili acted as the primary adhesin, the surface M1 protein clearly enhanced adhesion to tonsil, but surprisingly, had the opposite effect on adhesion to keratinocytes. These studies provide clear evidence that S. pyogenes pili display an adhesive specificity for clinically relevant human tissues and are likely to play a critical role in the initial stages of infection.
Collapse
Affiliation(s)
- Emily L Abbot
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang M, McDonald FM, Sturrock SS, Charnock SJ, Humphery-Smith I, Black GW. Group A streptococcus cell-associated pathogenic proteins as revealed by growth in hyaluronic acid-enriched media. Proteomics 2007; 7:1379-90. [PMID: 17407184 DOI: 10.1002/pmic.200600578] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Group A streptococcus (GAS), also know as Streptococcus pyogenes, is a human pathogen and can cause several fatal invasive diseases such as necrotising fasciitis, the so-called flesh-eating disease, and toxic shock syndrome. The destruction of connective tissue and the hyaluronic acid (HA) therein, is a key element of GAS pathogenesis. We therefore propagated GAS in HA-enriched growth media in an attempt to create a simple biological system that could reflect some elements of GAS pathogenesis. Our results show that several recognised virulence factors were up-regulated in HA-enriched media, including the M1 protein, a collagen-like surface protein and the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase, which has been shown to play important roles in streptococcal pathogenesis. Interestingly, two hypothetical proteins of unknown function were also up-regulated and detailed bioinformatics analysis showed that at least one of these hypothetical proteins is likely to be involved in pathogenesis. It was therefore concluded that this simple biological system provided a valuable tool for the identification of potential GAS virulence factors.
Collapse
Affiliation(s)
- Meng Zhang
- Biomolecular and Biomedical Research Centre, School of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK.
| | | | | | | | | | | |
Collapse
|
36
|
Rosch JW, Hsu FF, Caparon MG. Anionic lipids enriched at the ExPortal of Streptococcus pyogenes. J Bacteriol 2006; 189:801-6. [PMID: 17142392 PMCID: PMC1797331 DOI: 10.1128/jb.01549-06] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The ExPortal of Streptococcus pyogenes is a membrane microdomain dedicated to the secretion and folding of proteins. We investigated the lipid composition of the ExPortal by examining the distribution of anionic membrane phospholipids. Staining with 10-N-nonyl-acridine orange revealed a single microdomain enriched with an anionic phospholipid whose staining characteristics and behavior in a cardiolipin-deficient mutant were characteristic of phosphatidylglycerol. Furthermore, the location of the microdomain corresponded to the site of active protein secretion at the ExPortal. These results indicate that the ExPortal is an asymmetric lipid microdomain, whose enriched content of anionic phospholipids may play an important role in ExPortal organization and protein trafficking.
Collapse
Affiliation(s)
- Jason W Rosch
- Department of Molecular Microbiology, Washington University School of Medicine, Box 8230, 660 S. Euclid Ave. no. 8230, St. Louis, MO 63110-1093, USA
| | | | | |
Collapse
|
37
|
Timmer AM, Kristian SA, Datta V, Jeng A, Gillen CM, Walker MJ, Beall B, Nizet V. Serum opacity factor promotes group A streptococcal epithelial cell invasion and virulence. Mol Microbiol 2006; 62:15-25. [PMID: 16942605 DOI: 10.1111/j.1365-2958.2006.05337.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Serum opacity factor (SOF) is a bifunctional cell surface protein expressed by 40-50% of group A streptococcal (GAS) strains comprised of a C-terminal domain that binds fibronectin and an N-terminal domain that mediates opacification of mammalian sera. The sof gene was recently discovered to be cotranscribed in a two-gene operon with a gene encoding another fibronectin-binding protein, sfbX. We compared the ability of a SOF(+) wild-type serotype M49 GAS strain and isogenic mutants lacking SOF or SfbX to invade cultured HEp-2 human pharyngeal epithelial cells. Elimination of SOF led to a significant decrease in HEp-2 intracellular invasion while loss of SfbX had minimal effect. The hypoinvasive phenotype of the SOF(-) mutant could be restored upon complementation with the sof gene on a plasmid vector, and heterologous expression of sof49 in M1 GAS or Lactococcus lactis conferred marked increases in HEp-2 cell invasion. Studies using a mutant sof49 gene lacking the fibronectin-binding domain indicated that the N-terminal opacification domain of SOF contributes to HEp-2 invasion independent of the C-terminal fibronectin binding domain, findings corroborated by observations that a purified SOF N-terminal peptide could promote latex bead adherence to HEp-2 cells and inhibit GAS invasion of HEp-2 cells in a dose-dependent manner. Finally, the first in vivo studies to employ a single gene allelic replacement mutant of SOF demonstrate that this protein contributes to GAS virulence in a murine model of necrotizing skin infection.
Collapse
Affiliation(s)
- Anjuli M Timmer
- Department of Pediatrics, Division of Pharmacology and Drug Discovery, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Rosch JW, Caparon MG. The ExPortal: an organelle dedicated to the biogenesis of secreted proteins in Streptococcus pyogenes. Mol Microbiol 2006; 58:959-68. [PMID: 16262783 DOI: 10.1111/j.1365-2958.2005.04887.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Gram-positive pathogen Streptococcus pyogenes secretes proteins through the ExPortal, a unique single microdomain of the cellular membrane specialized to contain the Sec translocons. It has been proposed that the ExPortal functions as an organelle to promote the biogenesis of secreted proteins by coordinating interactions between nascent unfolded secretory proteins and membrane-associated chaperones. In this study we provide evidence to support this model. It was found that HtrA (DegP), a surface anchored accessory factor required for maturation of the secreted SpeB cysteine protease, was localized exclusively to the ExPortal. Furthermore, the ATP synthase beta subunit was not localized to the ExPortal, suggesting that retention is likely restricted to a specific subset of exported proteins. Mutations that disrupted the anchoring, but not the protease activity, of HtrA, also altered the maturation kinetics of SpeB demonstrating that localization to the ExPortal was important for HtrA function. These data indicate that the ExPortal provides a mechanism by which Gram-positive bacteria can coordinate protein secretion and subsequent biogenesis in the absence of a specialized protein-folding compartment.
Collapse
Affiliation(s)
- Jason W Rosch
- Department of Molecular Microbiology, Washington University School of Medicine, Box 8230, St. Louis, MO 63110-1093, USA
| | | |
Collapse
|
39
|
Jin H, Pancholi V. Identification and Biochemical Characterization of a Eukaryotic-type Serine/Threonine Kinase and its Cognate Phosphatase in Streptococcus pyogenes: Their Biological Functions and Substrate Identification. J Mol Biol 2006; 357:1351-72. [PMID: 16487973 DOI: 10.1016/j.jmb.2006.01.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2005] [Revised: 01/09/2006] [Accepted: 01/11/2006] [Indexed: 10/25/2022]
Abstract
A eukaryotic-type signaling system in group A Streptococcus (GAS) was identified and characterized. This system comprises primarily the products of two co-transcribed genes, a eukaryotic-type Ser/Thr kinase (SP-STK) and phosphatase (SP-STP) and their endogenous substrate histone-like protein (SP-HLP). Enzyme activities of SP-STK and SP-STP primarily depended on Mn(2+). The site on the substrate for reversible phosphorylation by these enzymes was found to be only the threonine residue. Using specific antibodies generated against these proteins, SP-STK was found to be membrane-associated with its N-terminal kinase domain facing the cytoplasm and its C-terminal repeat domain outside the membrane and cell-wall associated. Further, SP-STP, primarily a cytoplasmic protein, was found to be a major secretory protein of GAS and essential for bacterial survival. Three isogenic mutants, lacking either the entire SP-STK, or one of its two domains, were found displaying distinct pleiotropic effects on growth, colony morphology, cell division/septation, surface protein/virulence factor expression, bacterial ability to adhere to and invade human pharyngeal cells, and resist phagocytosis by human neutrophils. In addition to these properties, the ability of these three proteins to modulate the expression of the major virulence factors, the M protein and the capsule, indicates that these proteins are structurally and functionally distinct from the kinases and phosphatases described in other microorganisms and play a key role in GAS pathogenesis.
Collapse
Affiliation(s)
- Hong Jin
- Laboratory of Bacterial Pathogenesis, Department of Pathology, Ohio State University College of Medicine and Public Health, 288A, Tzgournis Medical Research Facility, 420 W 12th Avenue, Columbus, OH 43210-1214, USA
| | | |
Collapse
|
40
|
Boël G, Jin H, Pancholi V. Inhibition of cell surface export of group A streptococcal anchorless surface dehydrogenase affects bacterial adherence and antiphagocytic properties. Infect Immun 2005; 73:6237-48. [PMID: 16177295 PMCID: PMC1230963 DOI: 10.1128/iai.73.10.6237-6248.2005] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Surface dehydrogenase (SDH) is an anchorless, multifunctional protein displayed on the surfaces of group A Streptococcus (GAS) organisms. SDH is encoded by a single gene, sdh (gap or plr) that is essential for bacterial survival. Hence, the resulting nonfeasibility of creating a knockout mutant is a major limiting factor in studying its role in GAS pathogenesis. An insertion mutagenesis strategy was devised in which a nucleotide sequence encoding a hydrophobic tail of 12 amino acids ((337)IVLVGLVMLLLS(348)) was added at the 3' end of the sdh gene, successfully creating a viable mutant strain (M1-SDH(HBtail)). In this mutant strain, the SDH(HBtail) protein was not secreted in the medium but was retained in the cytoplasm and to some extent trapped within the cell wall. Hence, SDH(HBtail) was not displayed on the GAS surface. The mutant strain, M1-SDH(HBtail), grew at the same rate as the wild-type strain. The SDH(HBtail) protein displayed the same GAPDH activity as the wild-type SDH protein. Although the whole-cell extracts of the wild-type and mutant strains showed similar GAPDH activities, cell wall extracts of the mutant strain showed 5.5-fold less GAPDH activity than the wild-type strain. The mutant strain, M1-SDH(HBtail), bound significantly less human plasminogen, adhered poorly to human pharyngeal cells, and lost its innate antiphagocytic activity. These results indicate that the prevention of the cell surface export of SDH affects the virulence properties of GAS. The anchorless SDH protein, thus, is an important virulence factor.
Collapse
Affiliation(s)
- Grégory Boël
- Laboratory of Bacterial Pathogenesis, Public Health Research Institute at The International Center for Public Health, Newark, NJ 07103-3535, USA
| | | | | |
Collapse
|
41
|
Sherlock O, Vejborg RM, Klemm P. The TibA adhesin/invasin from enterotoxigenic Escherichia coli is self recognizing and induces bacterial aggregation and biofilm formation. Infect Immun 2005; 73:1954-63. [PMID: 15784535 PMCID: PMC1087433 DOI: 10.1128/iai.73.4.1954-1963.2005] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2004] [Revised: 10/20/2004] [Accepted: 11/22/2004] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli strains are responsible for many cases of gastrointestinal disease and represent a serious health problem worldwide. An essential step in the pathogenesis of such strains involves recognition and attachment to host intestinal surfaces. TibA is a potent bacterial adhesin associated with a number of enterotoxigenic E. coli strains and mediates bacterial attachment to a variety of human cells; additionally, it promotes invasion of such cells. This adhesin is a surface-displayed autotransporter protein and belongs to the exclusive group of bacterial glycoproteins; only the glycosylated form confers binding to and invasion of mammalian cells. Here we characterized TibA and showed that it possesses self-association characteristics and can mediate autoaggregation of E. coli cells. We demonstrated that intercellular TibA-TibA interaction is responsible for bacterial autoaggregation. Also, TibA expression significantly enhances biofilm formation by E. coli on abiotic surfaces.
Collapse
Affiliation(s)
- Orla Sherlock
- Centre for Biomedical Microbiology, BioCentrum-DTU, Bldg. 301, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | | | | |
Collapse
|
42
|
Zimmerlein B, Park HS, Li S, Podbielski A, Cleary PP. The M protein is dispensable for maturation of streptococcal cysteine protease SpeB. Infect Immun 2005; 73:859-64. [PMID: 15664926 PMCID: PMC546975 DOI: 10.1128/iai.73.2.859-864.2005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The streptococcal pyrogenic exotoxin B (SpeB) is an important virulence factor of group A streptococci (GAS) with cysteine protease activity. Maturation of SpeB to a proteolytically active form was suggested to be dependent on cell-wall-anchored M1 protein, the major surface protein of GAS (M. Collin and A. Olsen, Mol. Microbiol. 36:1306-1318, 2000). Collin and Olsen showed that mutant GAS strains expressing truncated M protein secrete a conformationally different form of unprocessed SpeB with no proteolytic activity. Alternatively, we hypothesized that a truncated M protein may interfere with processing of this secreted protease, and therefore we tested cysteine protease activity in genetically defined mutant strains that express either no M protein or membrane-anchored M protein with an in-frame deletion of the AB repeat region. Measurements of SpeB activity by cleavage of a substrate n-benzoyl-Pro-Phe-Arg-p-nitroanilide hydrochloride showed that the proteolytic activities in culture supernatants of both mutants were similar to those from the wild-type strain. In addition, Western blot analysis of culture supernatants showed that SpeB expression and processing to a mature form was unaffected by either deletion mutation. Therefore, we conclude that M protein is not required for maturation of the streptococcal cysteine protease SpeB.
Collapse
Affiliation(s)
- Björn Zimmerlein
- Department of Microbiology, University of Minnesota Medical School, 1460 Mayo Bldg., MMC196, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
43
|
Aziz RK, Ismail SA, Park HW, Kotb M. Post-proteomic identification of a novel phage-encoded streptodornase, Sda1, in invasive M1T1 Streptococcus pyogenes. Mol Microbiol 2004; 54:184-97. [PMID: 15458415 DOI: 10.1111/j.1365-2958.2004.04255.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The M1T1 strain remains the most frequently isolated strain from group A streptococcal (GAS) infection cases worldwide. We previously reported that M1T1 differs from the fully sequenced M1 SF370 strain. To better understand the reason for the persistence and increased virulence of M1T1, we analysed its secreted proteome and identified two virulence proteins that are not present in the sequenced M1 SF370 strain: streptococcal pyrogenic exotoxin A (SpeA) and a streptodornase D (SdaD) homologue. In the present study, we determined the nucleotide sequence of the M1T1 streptodornase and found that its deduced amino acid sequence is highly similar to other streptococcal streptodornases, and is most closely related to the SdaD of GAS strain M49. M1T1 Sda shares two highly conserved domains with several DNases and putative DNases in streptococci; however, it possesses a unique C-terminal amino acid sequence. Thus, we named the protein Sda1, and we detected the presence of the sda1 gene in 16 M1T1 clinical isolates. The cloned and expressed Sda1 degrades both streptococcal and mammalian DNA at physiological pH. Amino acid similarity analyses of known GAS deoxyribonucleases suggest that Sda1 may be a chimeric protein created through recombination events. Moreover, a natural mutation that resulted in longer Sda1 and SdaD as compared to other GAS DNases was found to confer increased activity on the protein. Analysis of the sequences flanking sda1 determined that it is carried by a prophage or a prophage-like element inserted in the tRNA-Ser gene of M1T1 GAS. Ongoing studies in our laboratory aim to determine the contribution of Sda1 to the virulence of this globally disseminated M1T1 strain.
Collapse
Affiliation(s)
- Ramy K Aziz
- Department of Molecular Sciences, University of Tennessee, Health Science Center, Memphis, TN, USA
| | | | | | | |
Collapse
|
44
|
Sherlock O, Schembri MA, Reisner A, Klemm P. Novel roles for the AIDA adhesin from diarrheagenic Escherichia coli: cell aggregation and biofilm formation. J Bacteriol 2004; 186:8058-65. [PMID: 15547278 PMCID: PMC529095 DOI: 10.1128/jb.186.23.8058-8065.2004] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Accepted: 08/13/2004] [Indexed: 11/20/2022] Open
Abstract
Diarrhea-causing Escherichia coli strains are responsible for numerous cases of gastrointestinal disease and constitute a serious health problem throughout the world. The ability to recognize and attach to host intestinal surfaces is an essential step in the pathogenesis of such strains. AIDA is a potent bacterial adhesin associated with some diarrheagenic E. coli strains. AIDA mediates bacterial attachment to a broad variety of human and other mammalian cells. It is a surface-displayed autotransporter protein and belongs to the selected group of bacterial glycoproteins; only the glycosylated form binds to mammalian cells. Here, we show that AIDA possesses self-association characteristics and can mediate autoaggregation of E. coli cells. We demonstrate that intercellular AIDA-AIDA interaction is responsible for bacterial autoaggregation. Interestingly, AIDA-expressing cells can interact with antigen 43 (Ag43)-expressing cells, which is indicative of an intercellular AIDA-Ag43 interaction. Additionally, AIDA expression dramatically enhances biofilm formation by E. coli on abiotic surfaces in flow chambers.
Collapse
Affiliation(s)
- Orla Sherlock
- Centre for Biomedical Microbiology, BioCentrum-DTU, Technical University of Denmark, DK-2800 Lyngby, Denmark
| | | | | | | |
Collapse
|
45
|
Lyon WR, Caparon MG. Role for serine protease HtrA (DegP) of Streptococcus pyogenes in the biogenesis of virulence factors SpeB and the hemolysin streptolysin S. Infect Immun 2004; 72:1618-25. [PMID: 14977969 PMCID: PMC356025 DOI: 10.1128/iai.72.3.1618-1625.2004] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The serine protease HtrA is involved in the folding and maturation of secreted proteins, as well as in the degradation of proteins that misfold during secretion. Depletion of HtrA has been shown to affect the sensitivity of many organisms to thermal and environmental stresses, as well as being essential for virulence in many pathogens. In the present study, we compared the behaviors of several different HtrA mutants of the gram-positive pathogen Streptococcus pyogenes (group A streptococcus). Consistent with prior reports, insertional inactivation of htrA, the gene that encodes HtrA, resulted in a mutant that grew poorly at 37 degrees C. However, an identical phenotype was observed when a similar polar insertion was placed immediately downstream of htrA in the streptococcal chromosome, suggesting that the growth defect of the insertion mutant was not a direct result of insertional inactivation of htrA. This conclusion was supported by the observation that a nonpolar deletion mutation of htrA did not produce the growth defect. However, this mutation did affect the production of several secreted virulence factors whose biogenesis requires extensive processing. For the SpeB cysteine protease, the loss of HtrA was associated with a failure to proteolytically process the zymogen to an active protease. For the streptolysin S hemolysin, a dramatic increase in hemolytic activity resulted from the depletion of HtrA. Interestingly, HtrA-deficient mutants were not attenuated in a murine model of subcutaneous infection. These data add to the growing body of information that implies an important role for HtrA in the biogenesis of secreted proteins in gram-positive bacteria.
Collapse
Affiliation(s)
- William R Lyon
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63110-1093, USA
| | | |
Collapse
|
46
|
Weineisen M, Sjöbring U, Fällman M, Andersson T. Streptococcal M5 protein prevents neutrophil phagocytosis by interfering with CD11b/CD18 receptor-mediated association and signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2004; 172:3798-807. [PMID: 15004185 DOI: 10.4049/jimmunol.172.6.3798] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Group A streptococci (GAS) are common human pathogens that express major surface-associated virulence factors designated M proteins. In this study, we explored directly the cellular mechanisms behind their supposed ability to prevent phagocytosis. Isolated human neutrophils killed an M-negative GAS mutant (DeltaM5), but not the wild-type parent strain (M5). After 3 h, 3-4 times as many DeltaM5 as M5 bacteria were associated with the neutrophils, and more DeltaM5 than M5 bacteria were ingested. However, there was no statistically significant difference between DeltaM5 and M5 bacteria in regard to the percentage of the neutrophil-associated bacteria that were ingested, indicating that M5 protein prevents an adhesion receptor-dependent association with neutrophils and not the phagocytic machinery per se. Different Abs against CD11b/CD18 (CR3) blocked adhesion and killing of DeltaM5 bacteria, whereas the blocking of two other complement receptors, CD11c/CD18 (CR4) and CD35 (CR1), did not. The CD11b/CD18-mediated killing of DeltaM5 bacteria resulted in protein tyrosine phosphorylations and Cdc42 activation. Furthermore, inhibition of CD11b/CD18 receptor engagement or tyrosine kinase activity blocked the DeltaM5-induced activation of Cdc42 as well as the killing of these bacteria. We conclude that M5 protein interferes with the CD11b/CD18-dependent association between GAS and neutrophils, and thereby blocks subsequent ingestion of the bacteria.
Collapse
Affiliation(s)
- Maria Weineisen
- Experimental Pathology, Lund University, Malmö University Hospital, Malmö, Sweden.
| | | | | | | |
Collapse
|
47
|
Tomich M, Mohr CD. Adherence and autoaggregation phenotypes of aBurkholderia cenocepaciacable pilus mutant. FEMS Microbiol Lett 2003; 228:287-97. [PMID: 14638436 DOI: 10.1016/s0378-1097(03)00785-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Cable pili are unique peritrichous adherence organelles expressed by certain strains of the opportunistic human pathogen Burkholderia cenocepacia. Cable pili have been proposed to facilitate binding to human epithelial cells and mucin, and may play a role in the ability of B. cenocepacia to colonise the respiratory tract of compromised hosts. In this study, a genetic approach was undertaken to assess the role of cable pili in mediating adherence as well as bacterial cell-cell interactions. The cblA gene, encoding the major pilin subunit, was insertionally inactivated, and the resulting mutant was shown to be blocked in CblA expression and in cable pilus morphogenesis. Although non-piliated, the cblA mutant was not defective in adherence to either porcine mucin or to cultured A549 human respiratory epithelial cells. Microscopic and flow cytometric analyses of B. cenocepacia cultures revealed that cable pilus expression facilitated the formation of diffuse cell networks, whereas disruption of cable pilus biogenesis enhanced autoaggregation and the formation of compact cell aggregates. Autoaggregation was observed both in culture and during B. cenocepacia infection of A549 epithelial cell monolayers. These findings indicate that cable pilus expression plays an important role in mediating B. cenocepacia cell-cell interactions, and that both cable pilus-dependent and cable pilus-independent mechanisms may contribute to B. cenocepacia adherence to cellular and acellular surfaces.
Collapse
Affiliation(s)
- Mladen Tomich
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN 55455-0312, USA
| | | |
Collapse
|
48
|
Gutekunst H, Eikmanns BJ, Reinscheid DJ. Analysis of RogB-controlled virulence mechanisms and gene repression in Streptococcus agalactiae. Infect Immun 2003; 71:5056-64. [PMID: 12933848 PMCID: PMC187362 DOI: 10.1128/iai.71.9.5056-5064.2003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae is the leading cause of bacterial sepsis and meningitis in neonates and also the causative agent of different serious infections in immunocompromised adults. The wide range of diseases that are caused by S. agalactiae suggests regulatory mechanisms that control the formation of specific virulence factors in these bacteria. The present study describes a gene from S. agalactiae, designated rogB, encoding a protein with significant similarity to members of the RofA-like protein (RALP) family of transcriptional regulators. Disruption of the rogB gene in the genome of S. agalactiae resulted in mutant strain RGB1, which was impaired in its ability to bind to fibrinogen and fibronectin. Mutant RGB1 also exhibited a reduced adherence to human epithelial cells but did not show an altered invasion of eukaryotic cells. By real-time PCR analysis, mutant RGB1 revealed an increased expression of the cpsA gene, encoding a regulator of capsule gene expression. However, strain RGB1 exhibited a reduced expression of the rogB gene and of two adjacent genes, encoding putative virulence factors in S. agalactiae. Furthermore, mutant RGB1 was impaired in the expression of the fbsA gene, coding for a fibrinogen receptor from S. agalactiae. The altered gene expression in mutant RGB1 could be restored by plasmid-mediated expression of rogB, confirming a RogB deficiency as the cause for the observed changes in virulence gene expression in S. agalactiae. Reporter gene studies with a promotorless luciferase gene fused to fbsA allowed a growth-dependent analysis of fbsA expression in S. agalactiae. These reporter gene studies also suggest that RogB exerts a positive effect on fbsA expression in S. agalactiae.
Collapse
Affiliation(s)
- Heike Gutekunst
- Department of Microbiology and Biotechnology, University of Ulm, D-89069 Ulm, Germany
| | | | | |
Collapse
|
49
|
Frick IM, Schmidtchen A, Sjöbring U. Interactions between M proteins of Streptococcus pyogenes and glycosaminoglycans promote bacterial adhesion to host cells. EUROPEAN JOURNAL OF BIOCHEMISTRY 2003; 270:2303-11. [PMID: 12752450 DOI: 10.1046/j.1432-1033.2003.03600.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Several microbial pathogens have been reported to interact with glycosaminoglycans (GAGs) on cell surfaces and in the extracellular matrix. Here we demonstrate that M protein, a major surface-expressed virulence factor of the human bacterial pathogen, Streptococcus pyogenes, mediates binding to various forms of GAGs. Hence, S. pyogenes strains expressing a large number of different types of M proteins bound to dermatan sulfate (DS), highly sulfated fractions of heparan sulfate (HS) and heparin, whereas strains deficient in M protein surface expression failed to interact with these GAGs. Soluble M protein bound DS directly and could also inhibit the interaction between DS and S. pyogenes. Experiments with M protein fragments and with streptococci expressing deletion constructs of M protein, showed that determinants located in the NH2-terminal part as well as in the C-repeat region of the streptococcal proteins are required for full binding to GAGs. Treatment with ABC-chondroitinase and HS lyase that specifically remove DS and HS chains from cell surfaces, resulted in significantly reduced adhesion of S. pyogenes bacteria to human epithelial cells and skin fibroblasts. Together with the finding that exogenous DS and HS could inhibit streptococcal adhesion, these data suggest that GAGs function as receptors in M protein-mediated adhesion of S. pyogenes.
Collapse
Affiliation(s)
- Inga-Maria Frick
- Department of Cell and Molecular Biology, Section for Molecular Pathogenesis, Lund University, Sweden.
| | | | | |
Collapse
|
50
|
Bisno AL, Brito MO, Collins CM. Molecular basis of group A streptococcal virulence. THE LANCET. INFECTIOUS DISEASES 2003; 3:191-200. [PMID: 12679262 DOI: 10.1016/s1473-3099(03)00576-0] [Citation(s) in RCA: 326] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The group A streptococcus (GAS) (Streptococcus pyogenes) is among the most common and versatile of human pathogens. It is responsible for a wide spectrum of human diseases, ranging from trivial to lethal. The advent of modern techniques of molecular biology has taught much about the organism's virulence, and the genomes of several GAS types have now been deciphered. Surface structures of GAS including a family of M proteins, the hyaluronic acid capsule, and fibronectin-binding proteins, allow the organism to adhere to, colonise, and invade human skin and mucus membranes under varying environmental conditions. M protein binds to complement control factors and other host proteins to prevent activation of the alternate complement pathway and thus evade phagocytosis and killing by polymorphonuclear leucocytes. Extracellular toxins, including superantigenic streptococcal pyrogenic exotoxins, contribute to tissue invasion and initiate the cytokine storm felt responsible for illnesses such as necrotising fasciitis and the highly lethal streptococcal toxic shock syndrome. Progress has been made in understanding the molecular epidemiology of acute rheumatic fever but less is understood about its basic pathogenesis. The improved understanding of GAS genetic regulation, structure, and function has opened exciting possibilities for developing safe and effective GAS vaccines. Studies directed towards achieving this long-sought goal are being aggressively pursued.
Collapse
Affiliation(s)
- A L Bisno
- Miami Veterans Affairs Medical Center, and the University of Miami School of Medicine, Miami, FL 33125, USA
| | | | | |
Collapse
|