1
|
Thacharodi A, Hassan S, Vithlani A, Ahmed T, Kavish S, Geli Blacknell NM, Alqahtani A, Pugazhendhi A. The burden of group A Streptococcus (GAS) infections: The challenge continues in the twenty-first century. iScience 2025; 28:111677. [PMID: 39877071 PMCID: PMC11773489 DOI: 10.1016/j.isci.2024.111677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
Streptococcus pyogenes is a Gram-positive bacterium, also known as Group A Streptococcus (GAS), that has become a significant threat to the healthcare system, infecting more than 18 million people and resulting in more than 500,000 deaths annually worldwide. GAS infection rates decreased gradually during the 20th century in Western countries, largely due to improved living conditions and access to antibiotics. However, post-COVID-19, the situation has led to a steep increase in GAS infection rates in Europe, the United States, Australia, and New Zealand, which triggers a global concern. GAS infections are normally moderate, with symptoms of fever, pharyngitis, and pyoderma; nevertheless, if left untreated or with continued exposure to GAS or with recurring infections it can result in fatal outcomes. GAS produces a variety of virulence factors and exotoxins that can lead to deadly infections such as necrotizing fasciitis, impetigo, cellulitis, pneumonia, empyema, streptococcal toxic shock syndrome, bacteremia, and puerperal sepsis. In addition, post-immune mediated disorders such as post-streptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease contribute to extremely high death rates in developing nations. Despite substantial research on GAS infections, it is still unclear what molecular pathways are responsible for their emergence and how to best manage them. This review thus provides insights into the most recent research on the pathogenesis, virulence, resistance, and host interaction mechanisms of GAS, as well as novel management options to assist scientific communities in combating GAS infections.
Collapse
Affiliation(s)
- Aswin Thacharodi
- Dr. Thacharodi’s Laboratories, Department of Research and Development, Puducherry 605005, India
| | - Saqib Hassan
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
- Future Leaders Mentoring Fellow, American Society for Microbiology, Washington 20036, USA
| | - Avadh Vithlani
- Senior Resident, Department of Pulmonary Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Tawfeeq Ahmed
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
| | - Sanjana Kavish
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu 600119, India
| | | | - Ali Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Arivalagan Pugazhendhi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
2
|
Shaw PK, Hayes AJ, Langton M, Berkhout A, Grimwood K, Davies MR, Walker MJ, Brouwer S. Clinical Snapshot of Group A Streptococcal Isolates from an Australian Tertiary Hospital. Pathogens 2024; 13:956. [PMID: 39599509 PMCID: PMC11597359 DOI: 10.3390/pathogens13110956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Streptococcus pyogenes (Group A Streptococcus, GAS) is a human-restricted pathogen that causes a wide range of diseases from pharyngitis and scarlet fever to more severe, invasive infections such as necrotising fasciitis and streptococcal toxic shock syndrome. There has been a global increase in both scarlet fever and invasive infections during the COVID-19 post-pandemic period. The aim of this study was the molecular characterisation of 17 invasive and non-invasive clinical non-emm1 GAS isolates from an Australian tertiary hospital collected between 2021 and 2022. Whole genome sequencing revealed a total of nine different GAS emm types with the most prevalent being emm22, emm12 and emm3 (each 3/17, 18%). Most isolates (14/17, 82%) carried at least one superantigen gene associated with contemporary scarlet fever outbreaks, and the carriage of these toxin genes was non-emm type specific. Several mutations within key regulatory genes were identified across the different GAS isolates, which may be linked to an increased expression of several virulence factors. This study from a single Australian centre provides a snapshot of non-emm1 GAS clinical isolates that are multiclonal and linked with distinct epidemiological markers commonly observed in high-income settings. These findings highlight the need for continual surveillance to monitor genetic markers that may drive future outbreaks.
Collapse
Affiliation(s)
- Phoebe K. Shaw
- Australian Infectious Diseases Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4067, Australia; (P.K.S.); (M.J.W.)
| | - Andrew J. Hayes
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; (A.J.H.); (M.R.D.)
| | - Maree Langton
- Gold Coast Laboratory, Pathology Queensland, Gold Coast, QLD 4215, Australia;
| | - Angela Berkhout
- Infection Management and Prevention Service, Queensland Specialist Immunisation Service, Children’s Health Queensland, South Brisbane, QLD 4101, Australia;
- Queensland Statewide Antimicrobial Stewardship Program, Department of Paediatrics, Royal Brisbane and Women’s Hospital, Gold Coast University Hospital, Southport, QLD 4215, Australia
| | - Keith Grimwood
- Gold Coast and Departments of Infectious Diseases and Paediatrics, Gold Coast Health, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4215, Australia;
| | - Mark R. Davies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia; (A.J.H.); (M.R.D.)
| | - Mark J. Walker
- Australian Infectious Diseases Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4067, Australia; (P.K.S.); (M.J.W.)
| | - Stephan Brouwer
- Australian Infectious Diseases Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4067, Australia; (P.K.S.); (M.J.W.)
| |
Collapse
|
3
|
Tomoyasu T, Tabata A, Nagamune H. Identification of mutations resulting in derepression of the intermedilysin gene by sequential mutagenesis of its promoter region in Streptococcus intermedius. FEMS Microbiol Lett 2024; 371:fnae063. [PMID: 39104214 DOI: 10.1093/femsle/fnae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/06/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024] Open
Abstract
Streptococcus intermedius secretes the human-specific cytolysin intermedilysin (ILY), a crucial factor in the pathogenicity of this bacterium. Previously, we reported that a lactose phosphotransferase repressor (LacR) represses ily expression, and that its mutation increases ILY production. Interestingly, UNS40, a strain isolated from a liver abscess, produces high levels of ILY despite the absence of mutations in the lacR promoter and coding regions. Our results showed that a G > A mutation at the -90th position from the transcription start point in the UNS40 ily promoter region increased hemolytic activity and decreased the binding ability to LacR. To elucidate the regions involved in the repression of ily expression, we generated mutant strains, in which point or deletion mutations were introduced into the ily promoter region, and then compared their hemolytic activity. Among the point mutations, -120 C > A and -90 G > A and their flanking mutations increased hemolytic activity. These results indicated that these mutations may increase the virulence of S. intermedius.
Collapse
Affiliation(s)
- Toshifumi Tomoyasu
- Department of Bioengineering, Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial and Social Sciences, Tokushima University Graduate School, 2-1 Minami-Jousanjima-Cho, Tokushima, Tokushima 770-8513, Japan
| | - Atsushi Tabata
- Department of Bioengineering, Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial and Social Sciences, Tokushima University Graduate School, 2-1 Minami-Jousanjima-Cho, Tokushima, Tokushima 770-8513, Japan
| | - Hideaki Nagamune
- Department of Bioengineering, Division of Bioscience and Bioindustry, Graduate School of Technology, Industrial and Social Sciences, Tokushima University Graduate School, 2-1 Minami-Jousanjima-Cho, Tokushima, Tokushima 770-8513, Japan
| |
Collapse
|
4
|
Sasagawa K, Domon H, Sakagami R, Hirayama S, Maekawa T, Isono T, Hiyoshi T, Tamura H, Takizawa F, Fukushima Y, Tabeta K, Terao Y. Matcha Green Tea Exhibits Bactericidal Activity against Streptococcus pneumoniae and Inhibits Functional Pneumolysin. Antibiotics (Basel) 2021; 10:antibiotics10121550. [PMID: 34943762 PMCID: PMC8698834 DOI: 10.3390/antibiotics10121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae is a causative pathogen of several human infectious diseases including community-acquired pneumonia. Pneumolysin (PLY), a pore-forming toxin, plays an important role in the pathogenesis of pneumococcal pneumonia. In recent years, the use of traditional natural substances for prevention has drawn attention because of the increasing antibacterial drug resistance of S. pneumoniae. According to some studies, green tea exhibits antibacterial and antitoxin activities. The polyphenols, namely the catechins epigallocatechin gallate (EGCG), epigallocatechin (EGC), epicatechin gallate (ECG), and epicatechin (EC) are largely responsible for these activities. Although matcha green tea provides more polyphenols than green tea infusions, its relationship with pneumococcal pneumonia remains unclear. In this study, we found that treatment with 20 mg/mL matcha supernatant exhibited significant antibacterial activity against S. pneumoniae regardless of antimicrobial resistance. In addition, the matcha supernatant suppressed PLY-mediated hemolysis and cytolysis by inhibiting PLY oligomerization. Moreover, the matcha supernatant and catechins inhibited PLY-mediated neutrophil death and the release of neutrophil elastase. These findings suggest that matcha green tea reduces the virulence of S. pneumoniae in vitro and may be a promising agent for the treatment of pneumococcal infections.
Collapse
Affiliation(s)
- Karin Sasagawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Rina Sakagami
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Satoru Hirayama
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Tomoki Maekawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Hikaru Tamura
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Fumio Takizawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Yoichi Fukushima
- Nestlé Japan Ltd., Wellness Communications, Tokyo 140-0002, Japan;
| | - Koichi Tabeta
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
- Correspondence: ; Tel.: +81-25-227-2838
| |
Collapse
|
5
|
Klar S, Poether DC, Reinert J, Hüttig N, Linsel G, Jäckel U. Application of impedance measurement to investigate in vitro inhalation toxicity of bacteria. J Occup Med Toxicol 2021; 16:32. [PMID: 34384434 PMCID: PMC8359036 DOI: 10.1186/s12995-021-00317-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Workers of agriculture and intensive life stock farming are exposed to highly contaminated workplaces. Bioaerosol exposures are suspected to trigger respiratory health effects of the workers. So far, risk evaluation of bioaerosols has been assessed through the infectivity of comprising biological agents that is classified in Europe by four risk groups according to the criteria of Directive 2000/54EC of the European Parliament. However, this directive additionally requires the risk assessment of allergenic and toxigenic effects without further elaboration. The aim of our study was to establish an in vitro screening system that is able to measure inhalative toxic effects of bacteria and their metabolites. METHODS In this study, we analyzed three bacterial toxins and five culture supernatants of selected bacteria with known toxicity as model agents exposed to the lung epithelial cell line NuLi-1. We used electrical cell-substrate impedance sensing (ECIS) method to monitor real-time cell changes and the viability test Prestoblue™. RESULTS We confirmed concentration dependent cytotoxic effects of the selected toxins in NuLi-1 cells over a period of up to 48 h. Each toxin resulted in a different but specific impedance profile over time according to their mode of action, whereas viability assay showed the metabolic activity of the cells at a chosen time point without revealing any information on their mode of action. Furthermore, dose-response-relationships were monitored. Tested model bacteria (Streptoccous pneumoniae, Acinetobacter radioresistens, Aerococcus viridans, Aeromonas hydrophila) reacted according to their expected toxicity except one bacterium (Enterococcus faecalis). The established assays revealed the concentration dependent onset and intensity of bacterial cytotoxicity and the viability of the cells at 24 h and 48 h exposure. CONCLUSION Impedance measurement and the viability assay Prestoblue™ in combination are suitable as sensitive screening methods to analyze toxic potential of bacteria and can therefor support the risk assessment of workplaces in terms of the directive 2000/54/EC.
Collapse
Affiliation(s)
- Stefanie Klar
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany.
| | - Dierk-Christoph Poether
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany
| | - Jessica Reinert
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany
| | - Nicole Hüttig
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany
| | - Gunter Linsel
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany
| | - Udo Jäckel
- Federal Institute for Occupational Safety and Health, Nöldnerstraße 40-42, 10317, Berlin, Germany
| |
Collapse
|
6
|
Brouwer S, Barnett TC, Ly D, Kasper KJ, De Oliveira DMP, Rivera-Hernandez T, Cork AJ, McIntyre L, Jespersen MG, Richter J, Schulz BL, Dougan G, Nizet V, Yuen KY, You Y, McCormick JK, Sanderson-Smith ML, Davies MR, Walker MJ. Prophage exotoxins enhance colonization fitness in epidemic scarlet fever-causing Streptococcus pyogenes. Nat Commun 2020; 11:5018. [PMID: 33024089 PMCID: PMC7538557 DOI: 10.1038/s41467-020-18700-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 09/01/2020] [Indexed: 02/03/2023] Open
Abstract
The re-emergence of scarlet fever poses a new global public health threat. The capacity of North-East Asian serotype M12 (emm12) Streptococcus pyogenes (group A Streptococcus, GAS) to cause scarlet fever has been linked epidemiologically to the presence of novel prophages, including prophage ΦHKU.vir encoding the secreted superantigens SSA and SpeC and the DNase Spd1. Here, we report the molecular characterization of ΦHKU.vir-encoded exotoxins. We demonstrate that streptolysin O (SLO)-induced glutathione efflux from host cellular stores is a previously unappreciated GAS virulence mechanism that promotes SSA release and activity, representing the first description of a thiol-activated bacterial superantigen. Spd1 is required for resistance to neutrophil killing. Investigating single, double and triple isogenic knockout mutants of the ΦHKU.vir-encoded exotoxins, we find that SpeC and Spd1 act synergistically to facilitate nasopharyngeal colonization in a mouse model. These results offer insight into the pathogenesis of scarlet fever-causing GAS mediated by prophage ΦHKU.vir exotoxins.
Collapse
Affiliation(s)
- Stephan Brouwer
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Timothy C Barnett
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Diane Ly
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Katherine J Kasper
- Department of Microbiology and Immunology and the Centre for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - David M P De Oliveira
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Amanda J Cork
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Liam McIntyre
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Magnus G Jespersen
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Johanna Richter
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Benjamin L Schulz
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
| | - Gordon Dougan
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, Hong Kong, China
| | - Yuanhai You
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing, 102206, China
| | - John K McCormick
- Department of Microbiology and Immunology and the Centre for Human Immunology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Martina L Sanderson-Smith
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Mark R Davies
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
7
|
Lucas R, Hadizamani Y, Gonzales J, Gorshkov B, Bodmer T, Berthiaume Y, Moehrlen U, Lode H, Huwer H, Hudel M, Mraheil MA, Toque HAF, Chakraborty T, Hamacher J. Impact of Bacterial Toxins in the Lungs. Toxins (Basel) 2020; 12:toxins12040223. [PMID: 32252376 PMCID: PMC7232160 DOI: 10.3390/toxins12040223] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
Bacterial toxins play a key role in the pathogenesis of lung disease. Based on their structural and functional properties, they employ various strategies to modulate lung barrier function and to impair host defense in order to promote infection. Although in general, these toxins target common cellular signaling pathways and host compartments, toxin- and cell-specific effects have also been reported. Toxins can affect resident pulmonary cells involved in alveolar fluid clearance (AFC) and barrier function through impairing vectorial Na+ transport and through cytoskeletal collapse, as such, destroying cell-cell adhesions. The resulting loss of alveolar-capillary barrier integrity and fluid clearance capacity will induce capillary leak and foster edema formation, which will in turn impair gas exchange and endanger the survival of the host. Toxins modulate or neutralize protective host cell mechanisms of both the innate and adaptive immunity response during chronic infection. In particular, toxins can either recruit or kill central players of the lung's innate immune responses to pathogenic attacks, i.e., alveolar macrophages (AMs) and neutrophils. Pulmonary disorders resulting from these toxin actions include, e.g., acute lung injury (ALI), the acute respiratory syndrome (ARDS), and severe pneumonia. When acute infection converts to persistence, i.e., colonization and chronic infection, lung diseases, such as bronchitis, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) can arise. The aim of this review is to discuss the impact of bacterial toxins in the lungs and the resulting outcomes for pathogenesis, their roles in promoting bacterial dissemination, and bacterial survival in disease progression.
Collapse
Affiliation(s)
- Rudolf Lucas
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| | - Yalda Hadizamani
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
| | - Joyce Gonzales
- Department of Medicine and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Thomas Bodmer
- Labormedizinisches Zentrum Dr. Risch, Waldeggstr. 37 CH-3097 Liebefeld, Switzerland;
| | - Yves Berthiaume
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
| | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital, Zürich, Steinwiesstrasse 75, CH-8032 Zürch, Switzerland;
| | - Hartmut Lode
- Insitut für klinische Pharmakologie, Charité, Universitätsklinikum Berlin, Reichsstrasse 2, D-14052 Berlin, Germany;
| | - Hanno Huwer
- Department of Cardiothoracic Surgery, Voelklingen Heart Center, 66333 Voelklingen/Saar, Germany;
| | - Martina Hudel
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Mobarak Abu Mraheil
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Haroldo Alfredo Flores Toque
- Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Trinad Chakraborty
- Justus-Liebig-University, Biomedical Research Centre Seltersberg, Schubertstr. 81, 35392 Giessen, Germany; (M.H.); (M.A.M.); (T.C.)
| | - Jürg Hamacher
- Lungen-und Atmungsstiftung, Bern, 3012 Bern, Switzerland;
- Pneumology, Clinic for General Internal Medicine, Lindenhofspital Bern, 3012 Bern, Switzerland
- Medical Clinic V-Pneumology, Allergology, Intensive Care Medicine and Environmental Medicine, Faculty of Medicine, Saarland University, University Medical Centre of the Saarland, D-66421 Homburg, Germany
- Institute for Clinical & Experimental Surgery, Faculty of Medicine, Saarland University, D-66421 Homburg, Germany
- Correspondence: (R.L.); (J.H.); Tel.: +41-31-300-35-00 (J.H.)
| |
Collapse
|
8
|
Matsue M, Ogura K, Sugiyama H, Miyoshi-Akiyama T, Takemori-Sakai Y, Iwata Y, Wada T, Okamoto S. Pathogenicity Characterization of Prevalent-Type Streptococcus dysgalactiae subsp. equisimilis Strains. Front Microbiol 2020; 11:97. [PMID: 32117127 PMCID: PMC7010647 DOI: 10.3389/fmicb.2020.00097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/17/2020] [Indexed: 12/26/2022] Open
Abstract
Streptococcus dysgalactiae subsp. equisimilis (SDSE) is an emerging human pathogen that causes severe invasive streptococcal diseases. Recent reports have shown that SDSE exhibits high pathogenicity with different mechanisms from that of Streptococcus pyogenes, although the two streptococci possess some common virulence factors such as streptolysin, streptokinase, and cell-binding proteins. To date, only a few studies have examined the variety of mechanisms expressing the pathogenicity of SDSE. Among nine SDSE clinical isolates sequenced in this study, we present in vitro and in vivo analyses of KNZ01 and KNZ03, whose emm and multilocus species types (MLSTs) are prevalent in Japan and other countries. For the comparison of pathogenicity, we also utilized the ATCC 12394 strain. The whole-genome analysis showed that KNZ03 and ATCC 12394 are categorized into an identical clonal complex by MLST and are phylogenetically close. However, the three strains exhibited different characteristics for pathogenicity in vitro; ATCC 12394 showed significant cytotoxicity to human keratinocytes and release of streptolysin O (SLO) compared to KNZ01 and KNZ03; KNZ03 exhibited significantly high hemolytic activity, but did not secrete SLO. KNZ01 and KNZ03 adhered to human keratinocytes at a higher rate than ATCC 12394; KNZ03 showed a higher rate of survival after a brief (30 min) incubation with human neutrophils compared to the other two strains; also, KNZ01 grew more rapidly in the presence of human serum. In vivo subcutaneous infection commonly resulted in ulcer formation in the three strains 7 days after infection. KNZ01-infected mice showed significant body weight loss 2 days after infection. Besides, on post-infection day 2, only KNZ01 remained in the cutaneous tissues of mice. Scanning electron microscopy analysis revealed that KNZ01 formed an extracellular structure (biofilm), which was probably composed of cell wall-anchoring proteins, in the presence of glucose and human serum. The extracellular structure of ATCC 12394 was also changed dramatically in response to culture conditions, whereas that of KNZ03 did not. Our study proposed that each SDSE strain possesses different virulence factors characteristics for mediating pathogenicity in humans.
Collapse
Affiliation(s)
- Miki Matsue
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Hironori Sugiyama
- Division of Instrumental Analysis, Engineering and Technology Department, Kanazawa University, Kanazawa, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Shinjuku, Japan
| | - Yukiko Takemori-Sakai
- Division of Clinical Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Division of Infection Control, Kanazawa University, Kanazawa, Japan
| | - Yasunori Iwata
- Division of Infection Control, Kanazawa University, Kanazawa, Japan.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Takashi Wada
- Division of Clinical Laboratory Medicine, Kanazawa University, Kanazawa, Japan.,Department of Nephrology and Laboratory Medicine, Kanazawa University, Kanazawa, Japan
| | - Shigefumi Okamoto
- Department of Clinical Laboratory Science, Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
9
|
Hydrogen peroxide release by bacteria suppresses inflammasome-dependent innate immunity. Nat Commun 2019; 10:3493. [PMID: 31375698 PMCID: PMC6677825 DOI: 10.1038/s41467-019-11169-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 06/26/2019] [Indexed: 12/30/2022] Open
Abstract
Hydrogen peroxide (H2O2) has a major function in host-microbial interactions. Although most studies have focused on the endogenous H2O2 produced by immune cells to kill microbes, bacteria can also produce H2O2. How microbial H2O2 influences the dynamics of host-microbial interactions is unclear. Here we show that H2O2 released by Streptococcus pneumoniae inhibits inflammasomes, key components of the innate immune system, contributing to the pathogen colonization of the host. We also show that the oral commensal H2O2-producing bacteria Streptococcus oralis can block inflammasome activation. This study uncovers an unexpected role of H2O2 in immune suppression and demonstrates how, through this mechanism, bacteria might restrain the immune system to co-exist with the host. The functions of microbial hydrogen peroxide (H2O2) in host-pathogen interactions are unclear. Here, Erttmann and Gekara show that H2O2 released by Streptococcus pneumoniae inhibits inflammasomes, and thereby contributes to the pathogen’s ability to colonize the host.
Collapse
|
10
|
Chirality Effect on Cholesterol Modulation of Protein Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 30649753 DOI: 10.1007/978-3-030-04278-3_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Cholesterol is a key steroidal, lipid biomolecule found abundantly in plasma membranes of eukaryotic cells. It is an important structural component of cellular membranes and regulates membrane fluidity and permeability. Cholesterol is also essential for normal functioning of key proteins including ion-channels, G protein-coupled receptors (GPCRs), membrane bound steroid receptors, and receptor kinases. It is thought that cholesterol exerts its actions via specific binding to chiral proteins and lipids as well as through non-specific physiochemical interactions. Distinguishing between the specific and the non-specific interactions can be difficult. Although much remains unclear, progress has been made in recent years by utilizing ent-cholesterol, the enantiomer of natural cholesterol (nat-cholesterol) as a probe. Ent-Cholesterol is the non-superimposable mirror image of nat-cholesterol and exhibits identical physiochemical properties as nat-cholesterol. Hence, if the biological effects of cholesterol result solely due to membrane effects, it is expected that there will be no difference between ent-cholesterol and nat-cholesterol. However, when direct binding with chiral proteins and lipids is involved, the enantiomer is expected to potentially elicit significantly different, measurable effects due to formation of diastereomeric complexes. In this chapter, we have reviewed the literature related to ent-cholesterol and its use as a probe for various biophysical and biological interactions of cholesterol.
Collapse
|
11
|
Lin L, Xu L, Lv W, Han L, Xiang Y, Fu L, Jin M, Zhou R, Chen H, Zhang A. An NLRP3 inflammasome-triggered cytokine storm contributes to Streptococcal toxic shock-like syndrome (STSLS). PLoS Pathog 2019; 15:e1007795. [PMID: 31170267 PMCID: PMC6553798 DOI: 10.1371/journal.ppat.1007795] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/28/2019] [Indexed: 02/01/2023] Open
Abstract
Infection with the Streptococcus suis (S. suis) epidemic strain can cause Streptococcal toxic shock-like syndrome (STSLS), which is characterized by a cytokine storm, dysfunction of multiple organs and a high incidence of mortality despite adequate treatment. Despite some progress concerning the contribution of the inflammatory response to STSLS, the precise mechanism underlying STSLS development remains elusive. Here, we use a murine model to demonstrate that caspase-1 activity is critical for STSLS development. Furthermore, we show that inflammasome activation by S. suis is mainly dependent on NLRP3 but not on NLRP1, AIM2 or NLRC4. The important role of NLRP3 activation in STSLS is further confirmed in vivo with the NLRP3 inhibitor MCC950 and nlrp3-knockout mice. By comparison of WT strain with isogenic strains with mutation of various virulence genes for inflammasome activation, Suilysin is essential for inflammasome activation, which is dependent on the membrane perforation activity to cause cytosolic K+ efflux. Moreover, the mutant strain msly (P353L) expressing mutagenic SLY without hemolytic activity was unable to activate the inflammasome and does not cause STSLS. In summary, we demonstrate that the high membrane perforation activity of the epidemic strain induces a high level of NLRP3 inflammasome activation, which is essential for the development of the cytokine storm and multi-organ dysfunction in STSLS and suggests NLRP3 inflammasome as an attractive target for the treatment of STSLS.
Collapse
Affiliation(s)
- Lan Lin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Lei Xu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Weihua Lv
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Li Han
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lei Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Meilin Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People’s Republic of China, Wuhan, Hubei, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| | - Anding Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People’s Republic of China, Wuhan, Hubei, China
| |
Collapse
|
12
|
Facey JA, Steele JR, Violi JP, Mitrovic SM, Cranfield C. An examination of microcystin-LR accumulation and toxicity using tethered bilayer lipid membranes (tBLMs). Toxicon 2018; 158:51-56. [PMID: 30521837 DOI: 10.1016/j.toxicon.2018.11.432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 01/23/2023]
Abstract
Microcystin-LR (MC-LR) is a potent cyanobacterial toxin responsible for animal and human poisonings worldwide. MC-LR is found in organisms throughout the foodweb, however there is conjecture regarding whether it biomagnifies. Few studies have investigated how MC-LR interacts with lipid membranes, a determinant of biomagnification potential. We tested whether 1 μM MC-LR irreversibly associates with lipid bilayers or causes the creation of pore defects upon short and long-term exposure. Using tethered bilayer lipid membranes (tBLMs), we observed an increase in membrane conduction in tBLMs, representing an interaction of microcystin-LR with the lipid bilayer and a change in membrane packing properties. However, there were minimal changes in membrane capacitance upon short and long-term exposure, and MC-LR exhibited a rapid off-rate. Upon 24 h exposure to the toxin, no lipophilic multimeric complexes were detected capable of altering the toxin's off-rate. There was no evidence of the creation of new pores. This study demonstrates that MC-LR does not irreversibly imbed itself into lipids membranes after short or long-term exposure and suggests MC-LR does not biomagnify through the food web via lipid storage.
Collapse
Affiliation(s)
- Jordan A Facey
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW, 2000, Australia.
| | - Joel R Steele
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW, 2000, Australia
| | - Jake P Violi
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW, 2000, Australia
| | - Simon M Mitrovic
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW, 2000, Australia
| | - Charles Cranfield
- School of Life Sciences, University of Technology Sydney, PO Box 123, Broadway, NSW, 2000, Australia
| |
Collapse
|
13
|
Vajjala A, Biswas D, Tay WH, Hanski E, Kline KA. Streptolysin-induced endoplasmic reticulum stress promotes group A Streptococcal host-associated biofilm formation and necrotising fasciitis. Cell Microbiol 2018; 21:e12956. [PMID: 30239106 DOI: 10.1111/cmi.12956] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/24/2022]
Abstract
Group A Streptococcus (GAS) is a human pathogen that causes infections ranging from mild to fulminant and life-threatening. Biofilms have been implicated in acute GAS soft-tissue infections such as necrotising fasciitis (NF). However, most in vitro models used to study GAS biofilms have been designed to mimic chronic infections and insufficiently recapitulate in vivo conditions along with the host-pathogen interactions that might influence biofilm formation. Here, we establish and characterise an in vitro model of GAS biofilm development on mammalian cells that simulates microcolony formation observed in a mouse model of human NF. We show that on mammalian cells, GAS forms dense aggregates that display hallmark biofilm characteristics including a 3D architecture and enhanced tolerance to antibiotics. In contrast to abiotic-grown biofilms, host-associated biofilms require the expression of secreted GAS streptolysins O and S (SLO, SLS) that induce endoplasmic reticulum (ER) stress in the host. In an in vivo mouse model, the streptolysin null mutant is attenuated in both microcolony formation and bacterial spread, but pretreatment of soft-tissue with an ER stressor restores the ability of the mutant to form wild-type-like microcolonies that disseminate throughout the soft tissue. Taken together, we have identified a new role of streptolysin-driven ER stress in GAS biofilm formation and NF disease progression.
Collapse
Affiliation(s)
- Anuradha Vajjala
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - Debabrata Biswas
- Cellular and Molecular Mechanisms of Inflammation, Campus for Research Excellence and Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore (NUS)-The Hebrew University of Jerusalem (HUJ), Singapore
| | - Wei Hong Tay
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate School, Nanyang Technological University, Singapore
| | - Emanuel Hanski
- Cellular and Molecular Mechanisms of Inflammation, Campus for Research Excellence and Technological Enterprise (CREATE), Department of Microbiology and Immunology, National University of Singapore (NUS)-The Hebrew University of Jerusalem (HUJ), Singapore.,Department of Microbiology and Molecular Genetics, Faculty of Medicine, The Institute for Medical Research, Israel-Canada (IMRIC), The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kimberly A Kline
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
14
|
Rakers L, Grill D, Matos AL, Wulff S, Wang D, Börgel J, Körsgen M, Arlinghaus HF, Galla HJ, Gerke V, Glorius F. Addressable Cholesterol Analogs for Live Imaging of Cellular Membranes. Cell Chem Biol 2018; 25:952-961.e12. [DOI: 10.1016/j.chembiol.2018.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/22/2018] [Accepted: 04/13/2018] [Indexed: 10/16/2022]
|
15
|
Bradshaw JL, Caballero AR, Bierdeman MA, Adams KV, Pipkins HR, Tang A, O'Callaghan RJ, McDaniel LS. Pseudomonas aeruginosa Protease IV Exacerbates Pneumococcal Pneumonia and Systemic Disease. mSphere 2018; 3:e00212-18. [PMID: 29720526 PMCID: PMC5932373 DOI: 10.1128/msphere.00212-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
Pneumonia is a pulmonary disease affecting people of all ages and is consistently a leading cause of childhood mortality and adult hospitalizations. Streptococcus pneumoniae and Pseudomonas aeruginosa are major lung pathogens commonly associated with community-acquired and nosocomial pneumonia. Additionally, mixed lung infections involving these bacterial pathogens are increasing in prevalence and are frequently more severe than single infections. The cooperative interactions of these two pathogens that impact pulmonary disease severity are understudied. A major secreted virulence factor of P. aeruginosa, protease IV (PIV), cleaves interleukin 22 (IL-22), a cytokine essential for maintaining innate mucosal defenses against extracellular pathogens. Here, we investigate the ability of PIV to augment the virulence of a pneumococcal strain with limited virulence, S. pneumoniae EF3030, in a C57BL/6 murine model of pneumonia. We demonstrate that pulmonary coinfection involving P. aeruginosa 103-29 and S. pneumoniae EF3030 results in pneumococcal bacteremia that is abrogated during pneumococcal coinfection with a PIV-deficient strain. Furthermore, intratracheal administration of exogenous PIV and EF3030 resulted in abundant immune cell infiltration into the lung with large abscess formation, as well as severe bacteremia leading to 100% mortality. Heat-inactivated PIV did not worsen pneumonia or reliably induce bacteremia, suggesting that the specific activity of PIV is required. Our studies also show that PIV depletes IL-22 in vivo Moreover, PIV-mediated enhancement of pneumonia and disease severity was dependent on the expression of pneumolysin (Ply), a prominent virulence factor of S. pneumoniae Altogether, we reveal that PIV and Ply additively potentiate pneumonia in a murine model of lung infection.IMPORTANCES. pneumoniae remains the leading cause of bacterial pneumonia despite widespread use of pneumococcal vaccines, forcing the necessity for appropriate treatment to control pneumococcal infections. Coinfections involving S. pneumoniae with other bacterial pathogens threaten antibiotic treatment strategies and disease outcomes. Currently, there is not an effective treatment for alveolar-capillary barrier dysfunction that precedes bacteremia. An understanding of the dynamics of host-pathogen interactions during single and mixed pulmonary infections could elucidate proper treatment strategies needed to prevent or reduce invasive disease. Antibiotic treatment decreases bacterial burden in the lung but also increases acute pathology due to cytotoxins released via antibiotic-induced bacterial lysis. Therefore, targeted therapeutics that inhibit or counteract the effects of bacterial proteases and toxins are needed in order to limit pathology and disease progression. This study identifies the cooperative effect of PIV and Ply, products of separate lung pathogens that additively alter the lung environment and facilitate invasive disease.
Collapse
Affiliation(s)
- Jessica L Bradshaw
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Armando R Caballero
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael A Bierdeman
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Kristen V Adams
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Haley R Pipkins
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Aihua Tang
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Richard J O'Callaghan
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Larry S McDaniel
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
16
|
Okshevsky M, Louw MG, Lamela EO, Nilsson M, Tolker‐Nielsen T, Meyer RL. A transposon mutant library of Bacillus cereus ATCC 10987 reveals novel genes required for biofilm formation and implicates motility as an important factor for pellicle-biofilm formation. Microbiologyopen 2018; 7:e00552. [PMID: 29164822 PMCID: PMC5911993 DOI: 10.1002/mbo3.552] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022] Open
Abstract
Bacillus cereus is one of the most common opportunistic pathogens causing foodborne illness, as well as a common source of contamination in the dairy industry. B. cereus can form robust biofilms on food processing surfaces, resulting in food contamination due to shedding of cells and spores. Despite the medical and industrial relevance of this species, the genetic basis of biofilm formation in B. cereus is not well studied. In order to identify genes required for biofilm formation in this bacterium, we created a library of 5000 + transposon mutants of the biofilm-forming strain B. cereusATCC 10987, using an unbiased mariner transposon approach. The mutant library was screened for the ability to form a pellicle biofilm at the air-media interface, as well as a submerged biofilm at the solid-media interface. A total of 91 genes were identified as essential for biofilm formation. These genes encode functions such as chemotaxis, amino acid metabolism and cellular repair mechanisms, and include numerous genes not previously known to be required for biofilm formation. Although the majority of disrupted genes are not directly responsible for motility, further investigations revealed that the vast majority of the biofilm-deficient mutants were also motility impaired. This observation implicates motility as a pivotal factor in the formation of a biofilm by B. cereus. These results expand our knowledge of the fundamental molecular mechanisms of biofilm formation by B. cereus.
Collapse
Affiliation(s)
- Mira Okshevsky
- Interdisciplinary Nanoscience CenterAarhus UniversityAarhusDenmark
| | | | | | - Martin Nilsson
- Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Tim Tolker‐Nielsen
- Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience CenterAarhus UniversityAarhusDenmark
- Department of BioscienceAarhus UniversityAarhusDenmark
| |
Collapse
|
17
|
Mucosal Infections and Invasive Potential of Nonencapsulated Streptococcus pneumoniae Are Enhanced by Oligopeptide Binding Proteins AliC and AliD. mBio 2018; 9:mBio.02097-17. [PMID: 29339428 PMCID: PMC5770551 DOI: 10.1128/mbio.02097-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nonencapsulated Streptococcus pneumoniae (NESp) is an emerging human pathogen that colonizes the nasopharynx and is associated with noninvasive diseases such as otitis media (OM), conjunctivitis, and nonbacteremic pneumonia. Since capsule expression was previously thought to be necessary for establishment of invasive pneumococcal disease (IPD), serotype-specific polysaccharide capsules are targeted by currently licensed pneumococcal vaccines. Yet, NESp expressing oligopeptide binding proteins AliC and AliD have been isolated during IPD. Thus, we hypothesize AliC and AliD are major NESp virulence determinants that facilitate persistence and development of IPD. Our study reveals that NESp expressing AliC and AliD have intensified virulence compared to isogenic mutants. Specifically, we demonstrate AliC and AliD enhance murine nasopharyngeal colonization and pulmonary infection and are required for OM in a chinchilla model. Furthermore, AliC and AliD increase pneumococcal survival in chinchilla whole blood and aid in resistance to killing by human leukocytes. Comparative proteome analysis revealed significant alterations in protein levels when AliC and AliD were absent. Virulence-associated proteins, including a pneumococcal surface protein C variant (CbpAC), were significantly downregulated, while starvation response indicators were upregulated in the double mutant relative to wild-type levels. We also reveal that differentially expressed CbpAC was essential for NESp adherence to epithelial cells, virulence during OM, reduction of C3b deposition on the NESp surface, and binding to nonspecific IgA. Altogether, the rise in NESp prevalence urges the need to understand how NESp establishes disease and persists in a host. This study highlights the roles of AliC, AliD, and CbpAC in the pathogenesis of NESp. Despite the effective, widespread use of licensed pneumococcal vaccines over many decades, pneumococcal infections remain a worldwide burden resulting in high morbidity and mortality. NESp subpopulations are rapidly rising in the wake of capsule-targeted vaccine strategies, yet there is very little knowledge on NESp pathogenic potential and virulence mechanisms. Although NESp lacks a protective capsule, NESp lineages expressing AliC and AliD have been associated with systemic infections. Furthermore, higher antibiotic resistance rates and transformation efficiencies associated with emerging NESp threaten treatment strategies needed to control pneumococcal infections and transmission. Elucidating how NESp survives within a host and establishes disease is necessary for development of broadened pneumococcal prevention methods. Our study identifies virulence determinants and host survival mechanisms employed by NESp with a high pathogenic potential. Moreover, our study also identifies virulence determinants shared by NESp and encapsulated strains that may serve as broad prevention and therapeutic targets.
Collapse
|
18
|
Degradation of nuclear Ubc9 induced by listeriolysin O is dependent on K + efflux. Biochem Biophys Res Commun 2017; 493:1115-1121. [PMID: 28911869 DOI: 10.1016/j.bbrc.2017.09.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/10/2017] [Indexed: 01/11/2023]
Abstract
Listeriolysin O (LLO) is a pore-forming toxin produced by L. monocytogenes, and is belonged to a protein family of cholesterol-dependent cytolysins (CDCs). Previous studies have demonstrated that LLO triggers Ubc9 degradation and disrupts host SUMOylation to facilitate bacterial infection. However, the underlying mechanism of Ubc9 degradation is unclear. Here we show that LLO-induced down-regulation of Ubc9 is independent of Ubc9-SUMO interaction, however, it may involve phosphorylation signaling. Additionally, LLO exerts its effects primarily on nuclear Ubc9 and this process is mediated by K+ efflux. Interestingly, for intracellular CDCs such as pneumolysin and suilysin, blockage of K+ efflux enhances degradation of nuclear Ubc9, suggesting that extracellular and intracellular pathogens may exploit different mechanisms to modulate host SUMOylation system. Furthermore, up-regulation of SUMOylation by stable expression of SUMO-1 or SUMO-2 shows a delay in membrane perforation by LLO, indicating that SUMO modification of host proteins may act at the frontline for the defense response against LLO. Taken together, our study provides insights to the understanding of host-pathogen interactions.
Collapse
|
19
|
Pectate Lyase Promoting Streptolysin O Expression in Escherichia coli and Strengthening Its Activity. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.14337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
20
|
Positive- and Negative-Control Pathways by Blood Components for Intermedilysin Production in Streptococcus intermedius. Infect Immun 2017; 85:IAI.00379-17. [PMID: 28607101 DOI: 10.1128/iai.00379-17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 11/20/2022] Open
Abstract
Streptococcus intermedius is an opportunistic bacterial pathogen secreting a human-specific cytolysin called intermedilysin (ILY) as a major pathogenic factor. This bacterium can degrade glycans into monosaccharides using two glycosidases, multisubstrate glycosidase A (MsgA) and neuraminidase (NanA). Here, we detected a stronger hemolytic activity mediated by ILY when S. intermedius PC574 was cultured in fetal bovine serum (FBS) than when it was grown in the standard culture medium. FBS-cultured cells also showed higher MsgA and NanA activity, although overproduction of ILY in FBS was undetectable in mutants nanA-null and msgA-null. Addition of purified MsgA and NanA to the FBS resulted in a release of 2.8 mM galactose and 4.3 mM N-acetylneuraminic acid; these sugar concentrations were sufficient to upregulate the expression of ILY, MsgA, and NanA. Conversely, when strain PC574 was cultured in human plasma, no similar increase in hemolytic activity was observed. Moreover, addition of human plasma to the culture in FBS appeared to inhibit the stimulatory effect of FBS on ILY, MsgA, and NanA, although there were individual differences among the plasma samples. We confirmed that human plasma contains immunoglobulins that can neutralize ILY, MsgA, and NanA activities. In addition, human plasma had a neutralizing effect on cytotoxicity of S. intermedius toward HepG2 cells in FBS, and a higher concentration of human plasma was necessary to reduce the cytotoxicity of an ILY-high-producing strain than an ILY-low-producing strain. Overall, our data show that blood contains factors that stimulate and inhibit ILY expression and activity, which may affect pathogenicity of S. intermedius.
Collapse
|
21
|
Sezgin E, Levental I, Mayor S, Eggeling C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol 2017; 18:361-374. [PMID: 28356571 PMCID: PMC5500228 DOI: 10.1038/nrm.2017.16] [Citation(s) in RCA: 1417] [Impact Index Per Article: 177.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cellular plasma membranes are laterally heterogeneous, featuring a variety of distinct subcompartments that differ in their biophysical properties and composition. A large number of studies have focused on understanding the basis for this heterogeneity and its physiological relevance. The membrane raft hypothesis formalized a physicochemical principle for a subtype of such lateral membrane heterogeneity, in which the preferential associations between cholesterol and saturated lipids drive the formation of relatively packed (or ordered) membrane domains that selectively recruit certain lipids and proteins. Recent studies have yielded new insights into this mechanism and its relevance in vivo, owing primarily to the development of improved biochemical and biophysical technologies.
Collapse
Affiliation(s)
- Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, 6431 Fannin Street, Houston, Texas 77030, USA
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bellary Road, Bangalore 560065, India
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| |
Collapse
|
22
|
Vdovikova S, Luhr M, Szalai P, Nygård Skalman L, Francis MK, Lundmark R, Engedal N, Johansson J, Wai SN. A Novel Role of Listeria monocytogenes Membrane Vesicles in Inhibition of Autophagy and Cell Death. Front Cell Infect Microbiol 2017; 7:154. [PMID: 28516064 PMCID: PMC5413512 DOI: 10.3389/fcimb.2017.00154] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/12/2017] [Indexed: 11/13/2022] Open
Abstract
Bacterial membrane vesicle (MV) production has been mainly studied in Gram-negative species. In this study, we show that Listeria monocytogenes, a Gram-positive pathogen that causes the food-borne illness listeriosis, produces MVs both in vitro and in vivo. We found that a major virulence factor, the pore-forming hemolysin listeriolysin O (LLO), is tightly associated with the MVs, where it resides in an oxidized, inactive state. Previous studies have shown that LLO may induce cell death and autophagy. To monitor possible effects of LLO and MVs on autophagy, we performed assays for LC3 lipidation and LDH sequestration as well as analysis by confocal microscopy of HEK293 cells expressing GFP-LC3. The results revealed that MVs alone did not affect autophagy whereas they effectively abrogated autophagy induced by pure LLO or by another pore-forming toxin from Vibrio cholerae, VCC. Moreover, Listeria monocytogenes MVs significantly decreased Torin1-stimulated macroautophagy. In addition, MVs protected against necrosis of HEK293 cells caused by the lytic action of LLO. We explored the mechanisms of LLO-induced autophagy and cell death and demonstrated that the protective effect of MVs involves an inhibition of LLO-induced pore formation resulting in inhibition of autophagy and the lytic action on eukaryotic cells. Further, we determined that these MVs help bacteria to survive inside eukaryotic cells (mouse embryonic fibroblasts). Taken together, these findings suggest that intracellular release of MVs from L. monocytogenes may represent a bacterial strategy to survive inside host cells, by its control of LLO activity and by avoidance of destruction from the autophagy system during infection.
Collapse
Affiliation(s)
- Svitlana Vdovikova
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Morten Luhr
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Paula Szalai
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Lars Nygård Skalman
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Monika K Francis
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden
| | - Richard Lundmark
- Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Department of Medical Biochemistry and Biophysics, Umeå UniversityUmeå, Sweden.,Department of Integrative Medical Biology, Umeå UniversityUmeå, Sweden
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of OsloOslo, Norway
| | - Jörgen Johansson
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| | - Sun N Wai
- Department of Molecular Biology, Umeå UniversityUmeå, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå UniversityUmeå, Sweden.,Umeå Centre for Microbial Research, Umeå UniversityUmeå, Sweden
| |
Collapse
|
23
|
Bandara M, Skehel JM, Kadioglu A, Collinson I, Nobbs AH, Blocker AJ, Jenkinson HF. The accessory Sec system (SecY2A2) in Streptococcus pneumoniae is involved in export of pneumolysin toxin, adhesion and biofilm formation. Microbes Infect 2017; 19:402-412. [PMID: 28456649 PMCID: PMC5526788 DOI: 10.1016/j.micinf.2017.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/27/2017] [Accepted: 04/17/2017] [Indexed: 01/04/2023]
Abstract
In Streptococcus pneumoniae TIGR4, genes encoding a SecY2A2 accessory Sec system are present within a locus encoding a serine-rich repeat surface protein PsrP. Mutant strains deleted in secA2 or psrP were deficient in biofilm formation, while the ΔsecA2 mutant was reduced in binding to airway epithelial cells. Cell wall protein (CWP) fractions from the ΔsecA2 mutant, but not from the ΔpsrP mutant, were reduced in haemolytic (pneumolysin) activity. Contact-dependent pneumolysin (Ply) activity of wild type TIGR4 cells was ten-fold greater than that of ΔsecA2 mutant cells suggesting that Ply was not active at the ΔsecA2 cell surface. Ply protein was found to be present in the CWP fraction from the ΔsecA2 mutant, but showed aberrant electrophoretic migration indicative of protein modification. Proteomic analyses led to the discovery that the ΔsecA2 mutant CWP fraction was deficient in two glycosidases as well as other enzymes involved in carbohydrate metabolism. Taken collectively the results suggest that positioning of Ply into the cell wall compartment in active form, together with glycosyl hydrolases and adhesins, requires a functional accessory Sec system.
Collapse
Affiliation(s)
- Mikaila Bandara
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK; School of Cellular & Molecular Medicine, University of Bristol, University Walk, Bristol, BS8 1TD, UK; School of Biochemistry, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - J Mark Skehel
- Biological Mass Spectrometry and Proteomics, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology & Immunology, Institute of Infection and Global Health, University of Liverpool, 8 West Derby Street, Liverpool L69 7BE, UK
| | - Ian Collinson
- School of Biochemistry, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Angela H Nobbs
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK
| | - Ariel J Blocker
- School of Cellular & Molecular Medicine, University of Bristol, University Walk, Bristol, BS8 1TD, UK; School of Biochemistry, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| | - Howard F Jenkinson
- School of Oral and Dental Sciences, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK.
| |
Collapse
|
24
|
Laborde RJ, Sanchez-Ferras O, Luzardo MC, Cruz-Leal Y, Fernández A, Mesa C, Oliver L, Canet L, Abreu-Butin L, Nogueira CV, Tejuca M, Pazos F, Álvarez C, Alonso ME, Longo-Maugéri IM, Starnbach MN, Higgins DE, Fernández LE, Lanio ME. Novel Adjuvant Based on the Pore-Forming Protein Sticholysin II Encapsulated into Liposomes Effectively Enhances the Antigen-Specific CTL-Mediated Immune Response. THE JOURNAL OF IMMUNOLOGY 2017; 198:2772-2784. [PMID: 28258198 DOI: 10.4049/jimmunol.1600310] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022]
Abstract
Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.
Collapse
Affiliation(s)
- Rady J Laborde
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Oraly Sanchez-Ferras
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María C Luzardo
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Yoelys Cruz-Leal
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Audry Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Circe Mesa
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liliana Oliver
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba
| | - Liem Canet
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Liane Abreu-Butin
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Catarina V Nogueira
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Mayra Tejuca
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Fabiola Pazos
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Carlos Álvarez
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - María E Alonso
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba
| | - Ieda M Longo-Maugéri
- Discipline of Immunology, Department of Microbiology, Immunology, and Parasitology, Paulista Medical School, Federal University of São Paulo, São Paulo 04023-900, Brazil; and
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Darren E Higgins
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Luis E Fernández
- Immunobiology Division, Center of Molecular Immunology, Havana 11600, Cuba;
| | - María E Lanio
- Center for Protein Studies, Faculty of Biology, University of Havana, Havana 10400, Cuba;
| |
Collapse
|
25
|
Chakrabarti RS, Ingham SA, Kozlitina J, Gay A, Cohen JC, Radhakrishnan A, Hobbs HH. Variability of cholesterol accessibility in human red blood cells measured using a bacterial cholesterol-binding toxin. eLife 2017; 6. [PMID: 28169829 PMCID: PMC5323040 DOI: 10.7554/elife.23355] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/28/2017] [Indexed: 01/07/2023] Open
Abstract
Cholesterol partitions into accessible and sequestered pools in cell membranes. Here, we describe a new assay using fluorescently-tagged anthrolysin O, a cholesterol-binding bacterial toxin, to measure accessible cholesterol in human red blood cells (RBCs). Accessible cholesterol levels were stable within individuals, but varied >10 fold among individuals. Significant variation was observed among ethnic groups (Blacks>Hispanics>Whites). Variation in accessibility of RBC cholesterol was unrelated to the cholesterol content of RBCs or plasma, but was associated with the phospholipid composition of the RBC membranes and with plasma triglyceride levels. Pronase treatment of RBCs only modestly altered cholesterol accessibility. Individuals on hemodialysis, who have an unexplained increase in atherosclerotic risk, had significantly higher RBC cholesterol accessibility. Our data indicate that RBC accessible cholesterol is a stable phenotype with significant inter-individual variability. Factors both intrinsic and extrinsic to the RBC contribute to variation in its accessibility. This assay provides a new tool to assess cholesterol homeostasis among tissues in humans. DOI:http://dx.doi.org/10.7554/eLife.23355.001
Collapse
Affiliation(s)
- Rima S Chakrabarti
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sally A Ingham
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Julia Kozlitina
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Austin Gay
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jonathan C Cohen
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Arun Radhakrishnan
- Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Helen H Hobbs
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States.,Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
26
|
Zhao X, Liu B, Liu S, Wang L, Wang J. Anticytotoxin Effects of Amentoflavone to Pneumolysin. Biol Pharm Bull 2017; 40:61-67. [PMID: 28049950 DOI: 10.1248/bpb.b16-00598] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pneumolysin (PLY) is a devastating bacterial protein toxin of Streptococcus pneumoniae that punctures the cytomembrane, leading to pathological reactions, such as cell disruption and inflammation. Drugs capable of closely impacting the toxin are considered advantageous in the treatment of bacterial infections. Amentoflavone (AMF) is a chemical substance extracted from traditional Chinese herbs. Previous studies have demonstrated that AMF has multiple pharmacological effects and mentioned without attenuating pneumolysin-mediated cytotoxicity. This work focuses on the influence of AMF on inhibitory hemolytic mechanisms. AMF interacts with the toxin at Ser254, Glu277, Arg359, and effectively weakens the oligomerization of wild-type PLY and provides considerable protection against pneumolysin-mediated human alveolar epithelial (A549) cell damage. The results of our study demonstrate that AMF could be a candidate against pneumolysin-related injury.
Collapse
Affiliation(s)
- Xiaoran Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University
| | | | | | | | | |
Collapse
|
27
|
Kaufmann SHE. EFIS lecture. Immune response to tuberculosis: How to control the most successful pathogen on earth. Immunol Lett 2016; 175:50-7. [PMID: 27181094 DOI: 10.1016/j.imlet.2016.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 12/11/2022]
Abstract
Tuberculosis (TB) remains a major health threat and general agreement exists that better control measures are needed. These include better diagnostics, drugs and vaccines. In particular, vaccines will be critical for better TB control. Based on knowledge about protective immunity against TB, a vaccine was created, VPM1002, which shows high protective efficacy and safety in experimental animal models. The vaccine has proven safe and immunogenic in human adults and neonates and is currently assessed in clinical trials in the context of HIV exposure. As a next step, a phase III efficacy trial in adults and a phase IIb efficacy trial in neonates are being planned. Biosignatures that differentially diagnose TB disease versus latent infection with high sensitivity and specificity have been designed. Most recently, a prognostic biosignature which predicts progression from latent infection to active TB has been identified. Biosignatures are not only of great value for improved diagnosis and prognosis of TB, they can also provide guidelines for better understanding of molecular mechanisms underlying disease. Accordingly, distinct biomarkers of diagnostic and prognostic value but of unknown biological function are being characterized functionally. In this way, deeper insights have been obtained on the role of type I interferon and of neutrophils in TB in experimental animal models of TB. In conclusion, clinical and basic research further supported by computational biology can complement each other in the pursuit of knowledge-based development of improved intervention measures for TB control.
Collapse
Affiliation(s)
- Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Department of Immunology, Berlin, Germany.
| |
Collapse
|
28
|
Zhao X, Li H, Wang J, Guo Y, Liu B, Deng X, Niu X. Verbascoside Alleviates Pneumococcal Pneumonia by Reducing Pneumolysin Oligomers. Mol Pharmacol 2016; 89:376-87. [PMID: 26700563 DOI: 10.1124/mol.115.100610] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/18/2015] [Indexed: 02/14/2025] Open
Abstract
Pneumolysin (PLY), an essential virulence factor of Streptococcus pneumoniae (pneumococcus), can penetrate the physical defenses of the host and possesses inflammatory properties. The vital role PLY plays in pneumococcus pathogenesis makes this virulence factor one of the most promising targets for the treatment of pneumococcal infection. Verbascoside (VBS) is an agent that does not exhibit bacteriostatic activity but has been shown to inhibit PLY-mediated cytotoxicity. The results from molecular dynamics simulations and mutational analysis indicated that VBS binds to the cleft between domains 3 and 4 of PLY, thereby blocking PLY's oligomerization and counteracting its hemolytic activity. Moreover, VBS can effectively alleviate PLY-mediated human alveolar epithelial (A549) cell injury, and treatment with VBS provides significant protection against lung damage and reduces mortality in a pneumococcal pneumonia murine model. Our results demonstrate that VBS is a strong candidate as a novel therapeutic in the treatment of Streptococcus pneumoniae infection.
Collapse
Affiliation(s)
- Xiaoran Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Hongen Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Jianfeng Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Yan Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Bowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.)
| | - Xuming Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.).
| | - Xiaodi Niu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China (X.Z., H.L., J.F., Y.G., B.L., X.D.); and Key Laboratory of Zoonosis, Ministry of Education, Department of Food Quality and Safety, Jilin University, Changchun, China (X.N.).
| |
Collapse
|
29
|
Barnett TC, Cole JN, Rivera-Hernandez T, Henningham A, Paton JC, Nizet V, Walker MJ. Streptococcal toxins: role in pathogenesis and disease. Cell Microbiol 2015; 17:1721-41. [PMID: 26433203 DOI: 10.1111/cmi.12531] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/13/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022]
Abstract
Group A Streptococcus (Streptococcus pyogenes), group B Streptococcus (Streptococcus agalactiae) and Streptococcus pneumoniae (pneumococcus) are host-adapted bacterial pathogens among the leading infectious causes of human morbidity and mortality. These microbes and related members of the genus Streptococcus produce an array of toxins that act against human cells or tissues, resulting in impaired immune responses and subversion of host physiological processes to benefit the invading microorganism. This toxin repertoire includes haemolysins, proteases, superantigens and other agents that ultimately enhance colonization and survival within the host and promote dissemination of the pathogen.
Collapse
Affiliation(s)
- Timothy C Barnett
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Jason N Cole
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Anna Henningham
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
30
|
Lee DH, Kim HR, Chung HY, Lim JG, Kim S, Kim SK, Ku HJ, Kim H, Ryu S, Choi SH, Lee JH. Complete genome sequence of Bacillus cereus FORC_005, a food-borne pathogen from the soy sauce braised fish-cake with quail-egg. Stand Genomic Sci 2015; 10:97. [PMID: 26566422 PMCID: PMC4642778 DOI: 10.1186/s40793-015-0094-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/31/2015] [Indexed: 11/10/2022] Open
Abstract
Due to abundant contamination in various foods, the pathogenesis of Bacillus cereus has been widely studied in physiological and molecular level. B. cereus FORC_005 was isolated from a Korean side dish, soy sauce braised fish-cake with quail-egg in South Korea. While 21 complete genome sequences of B. cereus has been announced to date, this strain was completely sequenced, analyzed, and compared with other complete genome sequences of B. cereus to elucidate the distinct pathogenic features of a strain isolated in South Korea. The genomic DNA containing a circular chromosome consists of 5,349,617-bp with a GC content of 35.29 %. It was predicted to have 5170 open reading frames, 106 tRNA genes, and 42 rRNA genes. Among the predicted ORFs, 3892 ORFs were annotated to encode functional proteins (75.28 %) and 1278 ORFs were predicted to encode hypothetical proteins (748 conserved and 530 non-conserved hypothetical proteins). This genome information of B. cereus FORC_005 would extend our understanding of its pathogenesis in genomic level for efficient control of its contamination in foods and further food poisoning.
Collapse
Affiliation(s)
- Dong-Hoon Lee
- Department of Food Science and Biotechnology, Institute of Life Science and Resources, Kyung Hee University, Yongin, Republic of Korea
| | - Hye Rim Kim
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Han Young Chung
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Jong Gyu Lim
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Suyeon Kim
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Se Keun Kim
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Hye-Jin Ku
- Department of Food Science and Biotechnology, Institute of Life Science and Resources, Kyung Hee University, Yongin, Republic of Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Sang Ho Choi
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, and Center for Food Safety and Toxicology, Seoul National University, Seoul, Republic of Korea
| | - Ju-Hoon Lee
- Department of Food Science and Biotechnology, Institute of Life Science and Resources, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
31
|
Marshall JE, Faraj BHA, Gingras AR, Lonnen R, Sheikh MA, El-Mezgueldi M, Moody PCE, Andrew PW, Wallis R. The Crystal Structure of Pneumolysin at 2.0 Å Resolution Reveals the Molecular Packing of the Pre-pore Complex. Sci Rep 2015; 5:13293. [PMID: 26333773 PMCID: PMC4558608 DOI: 10.1038/srep13293] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/16/2015] [Indexed: 11/09/2022] Open
Abstract
Pneumolysin is a cholesterol-dependent cytolysin (CDC) and virulence factor of Streptococcus pneumoniae. It kills cells by forming pores assembled from oligomeric rings in cholesterol-containing membranes. Cryo-EM has revealed the structures of the membrane-surface bound pre-pore and inserted-pore oligomers, however the molecular contacts that mediate these oligomers are unknown because high-resolution information is not available. Here we have determined the crystal structure of full-length pneumolysin at 1.98 Å resolution. In the structure, crystal contacts demonstrate the likely interactions that enable polymerisation on the cell membrane and the molecular packing of the pre-pore complex. The hemolytic activity is abrogated in mutants that disrupt these intermolecular contacts, highlighting their importance during pore formation. An additional crystal structure of the membrane-binding domain alone suggests that changes in the conformation of a tryptophan rich-loop at the base of the toxin promote monomer-monomer interactions upon membrane binding by creating new contacts. Notably, residues at the interface are conserved in other members of the CDC family, suggesting a common mechanism for pore and pre-pore assembly.
Collapse
Affiliation(s)
- Jamie E Marshall
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Bayan H A Faraj
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Alexandre R Gingras
- Department of Biochemistry, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Rana Lonnen
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Md Arif Sheikh
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Mohammed El-Mezgueldi
- Department of Biochemistry, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Peter C E Moody
- Department of Biochemistry, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Peter W Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| | - Russell Wallis
- Department of Infection, Immunity and Inflammation, University of Leicester, PO Box 138, Leicester, LE1 9HN UK.,Department of Biochemistry, University of Leicester, PO Box 138, Leicester, LE1 9HN UK
| |
Collapse
|
32
|
Hu DK, Liu Y, Li XY, Qu Y. In vitro expression of Streptococcus pneumoniae ply gene in human monocytes and pneumocytes. Eur J Med Res 2015; 20:52. [PMID: 25943628 PMCID: PMC4426643 DOI: 10.1186/s40001-015-0142-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 04/27/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae is one major cause of pneumonia in human and contains various virulence factors that contribute to pathogenesis of pneumococcal disease. This study investigated the role of pneumolysin, Ply, in facilitating S. pneumoniae invasion into the host blood stream. METHODS S. pneumoniae strains were isolated from clinical blood and sputum samples and confirmed by PCR. Expression of ply gene was assessed by infecting human monocytes and pneumocytes. RESULTS A total of 23 strains of S. pneumoniae isolated from blood (n = 11) and sputum (n = 12) along with S. pneumoniae ATCC49619 were used to infect human monocyte (THP-1) and Type II pneumocyte (A549) cell lines. All clinical strains of S. pneumoniae showed higher expression of ply mRNA than the American Type Culture Collection (ATCC) strain. Among the clinical strains, blood isolates showed higher expression of ply genes than sputum isolates, i.e., 2(1.5)- to 2(1.6)-folds in THP-1 and 2(0.4)- to 2(4.9)-folds in A549 cell lines. CONCLUSIONS The data from the current study demonstrated that ply gene of blood- and sputum-derived S. pneumoniae is differentially expressed in two different cell lines. Under survival pressure, ply is highly expressed in these two cell lines for blood-derived S. pneumoniae, indicating that ply gene may facilitate S. pneumoniae invasion into the host blood system.
Collapse
Affiliation(s)
- Da-Kang Hu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, 381# Zhongshan East Road, Taizhou, 318000, China.
| | - Yang Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanchang University, 17# Yong Wai Zheng Street, Nanchang, 330006, China.
| | - Xiang-Yang Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, 109# College West Road, Wenzhou, 325027, China.
| | - Ying Qu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, 381# Zhongshan East Road, Taizhou, 318000, China.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Tuberculosis (TB) remains a major health threat that will only be defeated by a combination of better drugs, diagnostics and vaccines. The only licensed TB vaccine, bacille Calmette-Guérin (BCG), protects against extrapulmonary TB in infants. RECENT FINDINGS Novel vaccine candidates that could protect against pulmonary TB either in TB naïve or in latent TB-infected healthy individuals have been developed and are currently being assessed in clinical trials. Subunit booster vaccines are either based on viral vectors expressing TB-specific antigens or on TB-protein antigens in adjuvants. Subunit vaccines are administered on top of BCG. Replacement vaccines for BCG are recombinant viable BCG or Mycobacterium tuberculosis. Several candidates are undergoing, or will soon start, phase IIb assessment for efficacy. The first vaccine candidate, MVA85A, to complete a phase IIb trial, unfortunately failed to show protection against TB in infants. Therapeutic vaccines composed of killed mycobacterial preparations target patients with complicated TB in adjunct to drug treatment. SUMMARY With increasing numbers of TB vaccine candidates in clinical trials, financial, regulatory and infrastructural issues arise, which would be best tackled by a global strategy. In addition, selection of the most promising vaccine candidates for further clinical development gains increasing importance.
Collapse
|
34
|
Streptolysin O and NAD-glycohydrolase prevent phagolysosome acidification and promote group A Streptococcus survival in macrophages. mBio 2014; 5:e01690-14. [PMID: 25227466 PMCID: PMC4172074 DOI: 10.1128/mbio.01690-14] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Group A Streptococcus (GAS, Streptococcus pyogenes) is an ongoing threat to human health as the agent of streptococcal pharyngitis, skin and soft tissue infections, and life-threatening conditions such as necrotizing fasciitis and streptococcal toxic shock syndrome. In animal models of infection, macrophages have been shown to contribute to host defense against GAS infection. However, as GAS can resist killing by macrophages in vitro and induce macrophage cell death, it has been suggested that GAS intracellular survival in macrophages may enable persistent infection. Using isogenic mutants, we now show that the GAS pore-forming toxin streptolysin O (SLO) and its cotoxin NAD-glycohydrolase (NADase) mediate GAS intracellular survival and cytotoxicity for macrophages. Unexpectedly, the two toxins did not inhibit fusion of GAS-containing phagosomes with lysosomes but rather prevented phagolysosome acidification. SLO served two essential functions, poration of the phagolysosomal membrane and translocation of NADase into the macrophage cytosol, both of which were necessary for maximal GAS intracellular survival. Whereas NADase delivery to epithelial cells is mediated by SLO secreted from GAS bound to the cell surface, in macrophages, the source of SLO and NADase is GAS contained within phagolysosomes. We found that transfer of NADase from the phagolysosome to the macrophage cytosol occurs not by simple diffusion through SLO pores but rather by a specific translocation mechanism that requires the N-terminal translocation domain of NADase. These results illuminate the mechanisms through which SLO and NADase enable GAS to defeat macrophage-mediated killing and provide new insight into the virulence of a major human pathogen. IMPORTANCE Macrophages constitute an important element of the innate immune response to mucosal pathogens. They ingest and kill microbes by phagocytosis and secrete inflammatory cytokines to recruit and activate other effector cells. Group A Streptococcus (GAS, Streptococcus pyogenes), an important cause of pharyngitis and invasive infections, has been shown to resist killing by macrophages. We find that GAS resistance to macrophage killing depends on the GAS pore-forming toxin streptolysin O (SLO) and its cotoxin NAD-glycohydrolase (NADase). GAS bacteria are internalized by macrophage phagocytosis but resist killing by secreting SLO, which damages the phagolysosome membrane, prevents phagolysosome acidification, and translocates NADase from the phagolysosome into the macrophage cytosol. NADase augments SLO-mediated cytotoxicity by depleting cellular energy stores. These findings may explain the nearly universal production of SLO by GAS clinical isolates and the association of NADase with the global spread of a GAS clone implicated in invasive infections.
Collapse
|
35
|
Abstract
The tuberculosis (TB) pandemic continues to rampage despite widespread use of the BCG (Bacillus Calmette-Guérin) vaccine. Novel vaccination strategies are urgently needed to arrest global transmission and prevent the uncontrolled development of multidrug-resistant forms of Mycobacterium tuberculosis. Over the last two decades, considerable progress has been made in the field of vaccine development with numerous innovative preclinical candidates and more than a dozen vaccines in clinical trials. These vaccines are developed either as boosters of the current BCG vaccine or as novel prime vaccines to replace BCG. Given the enormous prevalence of latent TB infection, vaccines that are protective on top of an already established infection remain a high priority and a significant scientific challenge. Here we discuss the current state of TB vaccine research and development, our understanding of the underlying immunology, and the requirements for an efficient TB vaccine.
Collapse
|
36
|
Glasser E, Rachamim T, Aharonovich D, Sher D. Hydra actinoporin-like toxin-1, an unusual hemolysin from the nematocyst venom of Hydra magnipapillata which belongs to an extended gene family. Toxicon 2014; 91:103-13. [PMID: 24768765 DOI: 10.1016/j.toxicon.2014.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/23/2014] [Accepted: 04/08/2014] [Indexed: 01/09/2023]
Abstract
Cnidarians rely on their nematocysts and the venom injected through these unique weaponry systems to catch prey and protect themselves from predators. The development and physiology of the nematocysts of Hydra magnipapillata, a classic model organism, have been intensively studied, yet the composition and biochemical activity of their venom components are mostly unknown. Here, we show that hydra actinoporin-like toxins (HALTs), which have previously been associated with Hydra nematocysts, belong to a multigene family comprising six genes, which have diverged from a single common ancestor. All six genes are expressed in a population of Hydra magnipapillata. When expressed recombinantly, HALT-1 (Δ-HYTX-Hma1a), an actinoporin-like protein found in the stenoteles (the main penetrating nematocysts used in prey capture), reveals hemolytic activity, albeit about two-thirds lower than that of the anemone actinoporin equinatoxin II (EqTII, Δ-AITX-Aeq1a). HALT-1 also differs from EqTII in the size of its pores, and likely does not utilize sphingomyelin as a membrane receptor. We describe features of the HALT-1 sequence which may contribute to this difference in activity, and speculate on the role of this unusual family of pore-forming toxins in the ecology of Hydra.
Collapse
Affiliation(s)
- Eliezra Glasser
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, 31905 Haifa, Israel
| | - Tamar Rachamim
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, 31905 Haifa, Israel
| | - Dikla Aharonovich
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, 31905 Haifa, Israel
| | - Daniel Sher
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, 31905 Haifa, Israel.
| |
Collapse
|
37
|
Kaufmann SH, Cotton MF, Eisele B, Gengenbacher M, Grode L, Hesseling AC, Walzl G. The BCG replacement vaccine VPM1002: from drawing board to clinical trial. Expert Rev Vaccines 2014; 13:619-30. [PMID: 24702486 DOI: 10.1586/14760584.2014.905746] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tuberculosis remains a major health threat and vaccines better than bacillus Calmette-Guérin (BCG) are urgently required. Here we describe our experience with a recombinant BCG expressing listeriolysin and deficient in urease. This potential replacement vaccine has demonstrated superior efficacy and safety over BCG in Mycobacterium tuberculosis aerosol-challenged mice and was safe in numerous animal models including immune-deficient mice, guinea pigs, rabbits and nonhuman primates. Phase I clinical trials in adults in Germany and South Africa have proven safety and a current Phase IIa trial is under way to assess immunogenicity and safety in its target population, newborns in a high tuberculosis incidence setting, with promising early results. Second-generation candidates are being developed to improve safety and efficacy.
Collapse
Affiliation(s)
- Stefan He Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Baruch M, Belotserkovsky I, Hertzog BB, Ravins M, Dov E, McIver KS, Le Breton YS, Zhou Y, Cheng CY, Chen CY, Hanski E. An extracellular bacterial pathogen modulates host metabolism to regulate its own sensing and proliferation. Cell 2014; 156:97-108. [PMID: 24439371 DOI: 10.1016/j.cell.2013.12.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 09/16/2013] [Accepted: 11/15/2013] [Indexed: 01/10/2023]
Abstract
Successful infection depends on the ability of the pathogen to gain nutrients from the host. The extracellular pathogenic bacterium group A Streptococcus (GAS) causes a vast array of human diseases. By using the quorum-sensing sil system as a reporter, we found that, during adherence to host cells, GAS delivers streptolysin toxins, creating endoplasmic reticulum stress. This, in turn, increases asparagine (ASN) synthetase expression and the production of ASN. The released ASN is sensed by the bacteria, altering the expression of ∼17% of GAS genes of which about one-third are dependent on the two-component system TrxSR. The expression of the streptolysin toxins is strongly upregulated, whereas genes linked to proliferation are downregulated in ASN absence. Asparaginase, a widely used chemotherapeutic agent, arrests GAS growth in human blood and blocks GAS proliferation in a mouse model of human bacteremia. These results delineate a pathogenic pathway and propose a therapeutic strategy against GAS infections.
Collapse
Affiliation(s)
- Moshe Baruch
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel
| | - Ilia Belotserkovsky
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel
| | - Baruch B Hertzog
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel
| | - Miriam Ravins
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel
| | - Eran Dov
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel
| | - Kevin S McIver
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institut, University of Maryland, College Park, MD 20742, USA
| | - Yoann S Le Breton
- Department of Cell Biology & Molecular Genetics and Maryland Pathogen Research Institut, University of Maryland, College Park, MD 20742, USA
| | - Yiting Zhou
- Mechanism of Inflammation Program, Center for Research Excellence & Technological Enterprise (CREATE), National University of Singapore and The Hebrew University of Jerusalem (HUJI), Singapore 138602, Singapore
| | - Catherine Youting Cheng
- Mechanism of Inflammation Program, Center for Research Excellence & Technological Enterprise (CREATE), National University of Singapore and The Hebrew University of Jerusalem (HUJI), Singapore 138602, Singapore
| | | | - Emanuel Hanski
- Department of Microbiology and Molecular Genetics, The Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem 91120, Israel; Mechanism of Inflammation Program, Center for Research Excellence & Technological Enterprise (CREATE), National University of Singapore and The Hebrew University of Jerusalem (HUJI), Singapore 138602, Singapore.
| |
Collapse
|
39
|
Progress in tuberculosis vaccine development and host-directed therapies--a state of the art review. THE LANCET RESPIRATORY MEDICINE 2014; 2:301-20. [PMID: 24717627 DOI: 10.1016/s2213-2600(14)70033-5] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tuberculosis continues to kill 1·4 million people annually. During the past 5 years, an alarming increase in the number of patients with multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis has been noted, particularly in eastern Europe, Asia, and southern Africa. Treatment outcomes with available treatment regimens for drug-resistant tuberculosis are poor. Although substantial progress in drug development for tuberculosis has been made, scientific progress towards development of interventions for prevention and improvement of drug treatment outcomes have lagged behind. Innovative interventions are therefore needed to combat the growing pandemic of multidrug-resistant and extensively drug-resistant tuberculosis. Novel adjunct treatments are needed to accomplish improved cure rates for multidrug-resistant and extensively drug-resistant tuberculosis. A novel, safe, widely applicable, and more effective vaccine against tuberculosis is also desperately sought to achieve disease control. The quest to develop a universally protective vaccine for tuberculosis continues. So far, research and development of tuberculosis vaccines has resulted in almost 20 candidates at different stages of the clinical trial pipeline. Host-directed therapies are now being developed to refocus the anti-Mycobacterium tuberculosis-directed immune responses towards the host; a strategy that could be especially beneficial for patients with multidrug-resistant tuberculosis or extensively drug-resistant tuberculosis. As we are running short of canonical tuberculosis drugs, more attention should be given to host-directed preventive and therapeutic intervention measures.
Collapse
|
40
|
|
41
|
LacR mutations are frequently observed in Streptococcus intermedius and are responsible for increased intermedilysin production and virulence. Infect Immun 2013; 81:3276-86. [PMID: 23798532 DOI: 10.1128/iai.00638-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus intermedius secretes a human-specific cytolysin, intermedilysin (ILY), which is considered to be the major virulence factor of this pathogen. We screened for a repressor of ily expression by using random gene disruption in a low-ILY-producing strain (PC574). Three independent high-ILY-producing colonies that had plasmid insertions within a gene that has high homology to lacR were isolated. Validation of these observations was achieved through disruption of lacR in strain PC574 with an erythromycin cassette, which also led to higher hemolytic activity, increased transcription of ily, and higher cytotoxicity against HepG2 cells, compared to the parental strain. The direct binding of LacR within the ily promoter region was shown by a biotinylated DNA probe pulldown assay, and the amount of ILY secreted into the culture supernatant by PC574 cells was increased by adding lactose or galactose to the medium as a carbon source. Furthermore, we examined lacR nucleotide sequences and the hemolytic activity of 50 strains isolated from clinical infections and 7 strains isolated from dental plaque. Of the 50 strains isolated from infections, 13 showed high ILY production, 11 of these 13 strains had one or more point mutations and/or an insertion mutation in LacR, and almost all mutations were associated with a marked decline in LacR function. These results strongly suggest that mutation in lacR is required for the overproduction of ILY, which is associated with an increase in pathogenicity of S. intermedius.
Collapse
|
42
|
The pore-forming haemolysins of bacillus cereus: a review. Toxins (Basel) 2013; 5:1119-39. [PMID: 23748204 PMCID: PMC3717773 DOI: 10.3390/toxins5061119] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 12/05/2022] Open
Abstract
The Bacillus cereus sensu lato group contains diverse Gram-positive spore-forming bacteria that can cause gastrointestinal diseases and severe eye infections in humans. They have also been incriminated in a multitude of other severe, and frequently fatal, clinical infections, such as osteomyelitis, septicaemia, pneumonia, liver abscess and meningitis, particularly in immuno-compromised patients and preterm neonates. The pathogenic properties of this organism are mediated by the synergistic effects of a number of virulence products that promote intestinal cell destruction and/or resistance to the host immune system. This review focuses on the pore-forming haemolysins produced by B. cereus: haemolysin I (cereolysin O), haemolysin II, haemolysin III and haemolysin IV (CytK). Haemolysin I belongs to the cholesterol-dependent cytolysin (CDC) family whose best known members are listeriolysin O and perfringolysin O, produced by L. monocytogenes and C. perfringens respectively. HlyII and CytK are oligomeric ß-barrel pore-forming toxins related to the α-toxin of S. aureus or the ß-toxin of C. perfringens. The structure of haemolysin III, the least characterized haemolytic toxin from the B. cereus, group has not yet been determined.
Collapse
|
43
|
Rennoll-Bankert KE, Dumler JS. Lessons from Anaplasma phagocytophilum: chromatin remodeling by bacterial effectors. Infect Disord Drug Targets 2013; 12:380-7. [PMID: 23082961 PMCID: PMC3664514 DOI: 10.2174/187152612804142242] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 08/27/2012] [Accepted: 08/27/2012] [Indexed: 02/06/2023]
Abstract
Bacterial pathogens can alter global host gene expression via histone modifications and chromatin remodeling in order to subvert host responses, including those involved with innate immunity, allowing for bacterial survival. Shigella flexneri, Listeria monocytogenes, Chlamydia trachomatis, and Anaplasma phagocytophilum express effector proteins that modify host histones and chromatin structure. A. phagocytophilum modulates granulocyte respiratory burst in part by dampening transcription of several key phagocyte oxidase genes. The A. phagocytophilum protein AnkA localizes to the myeloid cell nucleus where it binds AT-rich regions in the CYBB promoter and decreases its transcription. AT-rich regions of DNA are characteristic of matrix attachment regions (MARs) which are critical for chromatin structure and transcription. MAR-binding proteins, such as SATB1, interact with histone modifying enzymes resulting in altered gene expression. With A. phagocytophilum infection, histone deacetylase 1 (HDAC1) expression is increased and histone H3 acetylation is decreased at the CYBB promoter, suggesting a role for AnkA in altering host epigenetics and modulating gene transcription, at this, and perhaps other loci. This review will focus on how bacterial pathogens alter host epigenetics, by specifically examining A. phagocytophilum AnkA cis-regulation of CYBB transcription and epigenetic changes associated with infection.
Collapse
|
44
|
Nagel A, Prado M, Heitmann A, Tartz S, Jacobs T, Deschermeier C, Helm S, Stanway R, Heussler V. A new approach to generate a safe double-attenuated Plasmodium liver stage vaccine. Int J Parasitol 2013; 43:503-14. [DOI: 10.1016/j.ijpara.2013.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/11/2013] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
|
45
|
Kaufmann SH. Tuberculosis vaccines: Time to think about the next generation. Semin Immunol 2013; 25:172-81. [DOI: 10.1016/j.smim.2013.04.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 03/11/2013] [Accepted: 04/17/2013] [Indexed: 10/26/2022]
|
46
|
Gurel V, Lambert K, Page AE, Loynachan AT, Huges K, Timoney JF, Fettinger M, Horohov DW, McMichael J. Streptolysin-O/antibiotics adjunct therapy modulates site-specific expression of extracellular matrix and inflammatory genes in lungs of Rhodococcus equi infected foals. Vet Res Commun 2013; 37:145-54. [PMID: 23475766 DOI: 10.1007/s11259-013-9557-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2013] [Indexed: 11/30/2022]
Abstract
The addition of streptolysin-O (SLO) to the standard antibiotics regimen was shown to be superior to antibiotics alone after experimental infection of foals with Rhodoccocus equi (R. equi). The objective of this study is to investigate this response by determining the site-specific expression of extracellular matrix (ECM) and inflammatory response genes in biopsy samples taken from three distinct lung regions of the infected foals. Twenty-four foals were challenged by intrabronchial instillation of R. equi and assigned to four treatment groups: SLO/antibiotics adjunct therapy, antibiotics-only therapy (7.5 mg/kg clarithromycin and 5 mg/kg rifampin), SLO-only, and saline-only treatments. Treatments were administered twice daily for 16 days unless symptoms progressed to the point where the foals needed to be euthanized. Gene expressions were determined using custom-designed equine real-time qPCR arrays containing forty-eight genes from ECM remodeling and inflammation pathways. A non-parametric Wilcoxon signed-rank test for independent samples was applied to two pairs of time-matched comparison groups, SLO/antibiotics vs. antibiotics-only and SLO-only vs. saline-only, to document the significant differences in gene expressions within these groups. Several genes, MMP9, MMP2, TIMP2, COL1A1, COL12A1, ITGAL, ITGB1, FN1, CCL2, CCL3, CXCL9, TNFα, SMAD7, CD40, IL10, TGFB1, and TLR2, were significantly regulated compared to the unchallenged/untreated control foals. The results of this study demonstrate that enhancement of clinical responses by SLO is consistent with the changes in expression of critical genes in ECM remodeling and inflammatory response pathways.
Collapse
Affiliation(s)
- Volkan Gurel
- Beech Tree Labs Inc, 117 Chapman St, Providence, RI 02905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Targeted amino acid substitutions impair streptolysin O toxicity and group A Streptococcus virulence. mBio 2013; 4:e00387-12. [PMID: 23300245 PMCID: PMC3546560 DOI: 10.1128/mbio.00387-12] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptolysin O is a potent pore-forming toxin produced by group A Streptococcus. The aims of the present study were to dissect the relative contributions of different structural domains of the protein to hemolytic activity, to obtain a detoxified form of streptolysin O amenable to human vaccine formulation, and to investigate the role of streptolysin O-specific antibodies in protection against group A Streptococcus infection. On the basis of in silico structural predictions, we introduced two amino acid substitutions, one in the proline-rich domain 1 and the other in the conserved undecapeptide loop in domain 4. The resulting streptolysin O derivative showed no toxicity, was highly impaired in binding to eukaryotic cells, and was unable to form organized oligomeric structures on the cell surface. However, it was fully capable of conferring consistent protection in a murine model of group A Streptococcus infection. When we engineered a streptococcal strain to express the double-mutated streptolysin O, a drastic reduction in virulence as well as a diminished capacity to kill immune cells recruited at the infection site was observed. Furthermore, when mice immunized with the toxoid were challenged with the wild-type and mutant strains, protection only against the wild-type strain, not against the strain expressing the double-mutated streptolysin O, was obtained. We conclude that protection occurs by antibody-mediated neutralization of active toxin. We present a novel example of structural design of a vaccine antigen optimized for human vaccine use. Having previously demonstrated that immunization of mice with streptolysin O elicits a protective immune response against infection with group A Streptococcus strains of different serotypes, we developed in this study a double-mutated nontoxic derivative that represents a novel tool for the development of protective vaccine formulations against this important human pathogen. Furthermore, the innovative construction of an isogenic strain expressing a functionally inactive toxin and its use in infection and opsonophagocytosis experiments allowed us to investigate the mechanism by which streptolysin O mediates protection against group A Streptococcus. Finally, the ability of this toxin to directly attack and kill host immune cells during infection was studied in an air pouch model, which allowed parallel quantification of cellular recruitment, vitality, and cytokine release at the infection site.
Collapse
|
48
|
Fittipaldi N, Segura M, Grenier D, Gottschalk M. Virulence factors involved in the pathogenesis of the infection caused by the swine pathogen and zoonotic agent Streptococcus suis. Future Microbiol 2012; 7:259-79. [PMID: 22324994 DOI: 10.2217/fmb.11.149] [Citation(s) in RCA: 319] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Streptococcus suis is a major swine pathogen responsible for important economic losses to the swine industry worldwide. It is also an emerging zoonotic agent of meningitis and streptococcal toxic shock-like syndrome. Since the recent recognition of the high prevalence of S. suis human disease in southeast and east Asia, the interest of the scientific community in this pathogen has significantly increased. In the last few years, as a direct consequence of these intensified research efforts, large amounts of data on putative virulence factors have appeared in the literature. Although the presence of some proposed virulence factors does not necessarily define a S. suis strain as being virulent, several cell-associated or secreted factors are clearly important for the pathogenesis of the S. suis infection. In order to cause disease, S. suis must colonize the host, breach epithelial barriers, reach and survive in the bloodstream, invade different organs, and cause exaggerated inflammation. In this review, we discuss the potential contribution of different described S. suis virulence factors at each step of the pathogenesis of the infection. Finally, we briefly discuss other described virulence factors, virulence factor candidates and virulence markers for which a precise role at specific steps of the pathogenesis of the S. suis infection has not yet been clearly established.
Collapse
Affiliation(s)
- Nahuel Fittipaldi
- Groupe de Recherche sur les Maladies Infectieuses du Porc & Centre de Recherche en Infectiologie Porcine, Faculté de médecine vétérinaire, Université de Montréal, 3200 rue Sicotte, CP5000, St-Hyacinthe, Quebec, J2S 7C6, Canada
| | | | | | | |
Collapse
|
49
|
Comparative pathogenomics of bacteria causing infectious diseases in fish. INTERNATIONAL JOURNAL OF EVOLUTIONARY BIOLOGY 2012; 2012:457264. [PMID: 22675651 PMCID: PMC3364575 DOI: 10.1155/2012/457264] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 03/20/2012] [Indexed: 11/18/2022]
Abstract
Fish living in the wild as well as reared in the aquaculture facilities are susceptible to infectious diseases caused by a phylogenetically diverse collection of bacterial pathogens. Control and treatment options using vaccines and drugs are either inadequate, inefficient, or impracticable. The classical approach in studying fish bacterial pathogens has been looking at individual or few virulence factors. Recently, genome sequencing of a number of bacterial fish pathogens has tremendously increased our understanding of the biology, host adaptation, and virulence factors of these important pathogens. This paper attempts to compile the scattered literature on genome sequence information of fish pathogenic bacteria published and available to date. The genome sequencing has uncovered several complex adaptive evolutionary strategies mediated by horizontal gene transfer, insertion sequence elements, mutations and prophage sequences operating in fish pathogens, and how their genomes evolved from generalist environmental strains to highly virulent obligatory pathogens. In addition, the comparative genomics has allowed the identification of unique pathogen-specific gene clusters. The paper focuses on the comparative analysis of the virulogenomes of important fish bacterial pathogens, and the genes involved in their evolutionary adaptation to different ecological niches. The paper also proposes some new directions on finding novel vaccine and chemotherapeutic targets in the genomes of bacterial pathogens of fish.
Collapse
|
50
|
Abstract
In this review we discuss recent progress in the development, testing, and clinical evaluation of new vaccines against tuberculosis (TB). Over the last 20 years, tremendous progress has been made in TB vaccine research and development: from a pipeline virtually empty of new TB candidate vaccines in the early 1990s, to an era in which a dozen novel TB vaccine candidates have been and are being evaluated in human clinical trials. In addition, innovative approaches are being pursued to further improve existing vaccines, as well as discover new ones. Thus, there is good reason for optimism in the field of TB vaccines that it will be possible to develop better vaccines than BCG, which is still the only vaccine available against TB.
Collapse
Affiliation(s)
- Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|