1
|
Song M, Sun J, Lv K, Li J, Shi J, Xu Y. A comprehensive review of pathology and treatment of staphylococcus aureus osteomyelitis. Clin Exp Med 2025; 25:131. [PMID: 40299136 PMCID: PMC12040984 DOI: 10.1007/s10238-025-01595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/10/2025] [Indexed: 04/30/2025]
Abstract
Osteomyelitis (OM) is an inflammation of the bone and bone marrow triggered by infectious pathogens which may induce progressive bone destruction. The majority of OM cases, especially the chronic OM cases, are induced by the most prevalent and devastating pathogen Staphylococcus aureus (S. aureus), partially due to its resistance mechanisms against the immune system and antibiotic therapies. Regarding the high rate of morbidity and recurrence in patients, it is pivotal to elucidate underlying mechanisms that how S. aureus enter and survive in hosts. The accumulated discoveries have identified multiple distinct strategies associated with chronicity and recurrence include biofilm development, small colony variants (SCVs), staphylococcus abscess communities (SACs), the osteocyte lacuno-canalicular network invasion (OLCN) of cortical bones, and S. aureus protein A (SpA). Unfortunately, little clinical progress has been achieved for the diagnosis and therapeutic treatment for OM patients, indicating that numerous questions remain to be solved. Therefore, we still have a long way to obtain the clear elucidation of the host-pathogen interactions which could be applied for clinical treatment of OM. In this review, we provide insights of current knowledge about how S. aureus evades immune eradication and remains persistent in hosts with recent discoveries. The common and novel treatment strategies for OM are also described. The purpose of this review is to have in-dept understanding of S. aureus OM and bring new perspectives to therapeutic fields which may be translated to the clinic.
Collapse
Affiliation(s)
- Muguo Song
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Sun
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Kehan Lv
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Junyi Li
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Shi
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| | - Yongqing Xu
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| |
Collapse
|
2
|
Torpy H, Chau TH, Chatterjee S, Chernykh A, Torpy DJ, Meyer EJ, Thaysen-Andersen M. Impact of different pathogen classes on the serum N-glycome in septic shock. BBA ADVANCES 2025; 7:100138. [PMID: 39877544 PMCID: PMC11773048 DOI: 10.1016/j.bbadva.2025.100138] [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: 11/24/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
The morbidity and mortality of sepsis remain high. Clinicians lack effective markers to rapidly diagnose sepsis and identify the underlying pathogen infection particularly for patients with candidaemia or cases of culture-negative sepsis where culture-based diagnostics are inadequate. In our search for new lines of potential sepsis biomarkers, we here explore the impact of various classes of infectious agents on the serum N-glycome in a septic shock cohort. Comparative N-glycomics was performed on sera collected from 49 septic shock patients infected with viral (n = 9), bacterial (n = 37) or fungal (n = 3) pathogens using an established PGC-LC-MS/MS method. Aberrant serum N-glycosylation features were observed in patients with fungal infection relative to the other infection sub-groups including i) altered expression of prominent α2,6-sialylated biantennary N-glycan isomers, ii) elevated levels of IgG-type N-glycosylation and iii) a global shift in the serum N-glycome involving altered glycan type distribution and considerable changes in core fucosylation and α2,6-sialylation. Septic shock patients infected with bacterial and viral pathogens exhibited similar global serum N-glycome features and therefore could not be stratified based on their serum N-glycosylation. Subtle and less consistent serum N-glycome differences were observed between septic shock patients infected with different bacterial pathogens. In conclusion, our study has tested the impact of different pathogen classes on the serum N-glycome in a septic shock cohort, and reports that fungal infection impacts the host serum N-glycome differently compared to bacterial or viral infections thus potentially opening avenues for glycan-based biomarkers to better diagnose patients with candidaemia.
Collapse
Affiliation(s)
- Helena Torpy
- Infectious Diseases Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia
| | - The Huong Chau
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | | | - Anastasia Chernykh
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - David J. Torpy
- School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Emily J. Meyer
- School of Medicine, University of Adelaide, Adelaide, SA 5000, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
- Endocrine and Diabetes Services, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia
| | - Morten Thaysen-Andersen
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Institute for Glyco-core Research, Nagoya University, Nagoya, Aichi 464-0813, Japan
| |
Collapse
|
3
|
Kates SL, Owen JR, Xie C, Ren Y, Muthukrishnan G, Schwarz EM. Vaccines: Do they have a role in orthopedic trauma? Injury 2024; 55 Suppl 6:111631. [PMID: 39482036 DOI: 10.1016/j.injury.2024.111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 11/03/2024]
Abstract
Although vaccines have been hailed as one of the greatest advances in medicine based on their unparalleled cost-effectiveness in eradicating life-threatening infectious diseases, their role in orthopedic trauma-related infections is unclear. This is largely because vaccines are primarily made against pathogens that cause communicable diseases rather than opportunistic infections secondary to trauma, and most successful vaccines are against viruses rather than biofilm forming bacteria. Nonetheless, the tremendous costs to patients and healthcare systems warrant orthopedic trauma vaccine research, which has been a focal topic in recent international consensus meetings on musculoskeletal infection. This subject was also covered at the 2023 Osteosynthesis and Trauma Care Foundation (OTCF) meeting in Rome, Italy, and the purpose of this supplement article is to (1) highlight the osteoimmunology, animal models, translational research and clinical pilots that were discussed, (2) the proposed future directions that could lead to diagnostics and prognostics that are critically needed for evidence-based decision making, and (3) vaccines and passive-immunization strategies that could potentially be utilized to treat patients with orthopedic infections.
Collapse
Affiliation(s)
- Stephen L Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - John R Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Youliang Ren
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
4
|
Hendriks A, Kerkman PF, Varkila MRJ, Haitsma Mulier JLG, Ali S, Ten Doesschate T, van der Vaart TW, de Haas CJC, Aerts PC, Cremer OL, Bonten MJM, Nizet V, Liu GY, Codée JDC, Rooijakkers SHM, van Strijp JAG, van Sorge NM. Glycan-specific IgM is critical for human immunity to Staphylococcus aureus. Cell Rep Med 2024; 5:101734. [PMID: 39293400 PMCID: PMC11525025 DOI: 10.1016/j.xcrm.2024.101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/18/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
Staphylococcus aureus is a major human pathogen, yet the immune factors that protect against infection remain elusive. High titers of opsonic IgG antibodies, achieved in preclinical animal immunization studies, have consistently failed to provide protection in humans. Here, we investigate antibody responses to the conserved S. aureus surface glycan wall teichoic acid (WTA) and detect the presence of WTA-specific IgM and IgG antibodies in the plasma of healthy individuals. Functionally, WTA-specific IgM outperforms IgG in opsonophagocytic killing of S. aureus and protects against disseminated S. aureus bacteremia through passive immunization. In a clinical setting, patients with S. aureus bacteremia have significantly lower WTA-specific IgM but similar IgG levels compared to healthy controls. Importantly, low WTA-IgM levels correlate with disease mortality and impaired bacterial opsonization. Our findings may guide risk stratification of hospitalized patients and inform future design of antibody-based therapies and vaccines against serious S. aureus infection.
Collapse
Affiliation(s)
- Astrid Hendriks
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Priscilla F Kerkman
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Meri R J Varkila
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jelle L G Haitsma Mulier
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Sara Ali
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Thijs Ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Thomas W van der Vaart
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Carla J C de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Piet C Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marc J M Bonten
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - George Y Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Suzan H M Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands; Netherlands Reference Center for Bacterial Meningitis, Amsterdam UMC, location AMC, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Marín-Romero A, Pernagallo S. A comprehensive review of Dynamic Chemical Labelling on Luminex xMAP technology: a journey towards Drug-Induced Liver Injury testing. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:6139-6149. [PMID: 37965948 DOI: 10.1039/d3ay01481a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Drug-Induced Liver Injury (DILI) is a grave global adverse event that can result in fatal consequences, causing drug failures, market withdrawals, and regulatory warnings, leading to substantial financial losses. The early detection of DILI remains a significant challenge in global healthcare. Although circulating microRNAs (miRs) show promise as clinical biomarkers for DILI, the current analytical methods for their measurement are insufficient. There is a pressing need for rapid and reliable miR detection methods that eliminate the need for nucleic acid extraction and PCR-based amplification. This review highlights recent advancements achieved by integrating Dynamic Chemical Labelling (DCL) with Luminex xMAP technology. This powerful combination has resulted in groundbreaking bead-based assays that allow (1) the direct, multiplex detection of miRs, and (2) the simultaneous testing of miR and protein biomarkers. This triple capability enables a comprehensive assessment that significantly enhances the detection and analysis of crucial biomarkers, thus improving the understanding and diagnosis of DILI. In conclusion, this review offers valuable insights into the capabilities and potential applications of these groundbreaking assays in DILI research, as well as their potential use in other diagnostic and research domains that require direct or multiplex analysis of miRs or analysis of miRs in combination with proteins.
Collapse
Affiliation(s)
- Antonio Marín-Romero
- DESTINA Genomica S.L., Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Innovación 1, Edificio BIC, Armilla, Granada 18100, Spain.
| | - Salvatore Pernagallo
- DESTINA Genomica S.L., Parque Tecnológico Ciencias de la Salud (PTS), Avenida de la Innovación 1, Edificio BIC, Armilla, Granada 18100, Spain.
| |
Collapse
|
6
|
Sohail MU, Mashood F, Oberbach A, Chennakkandathil S, Schmidt F. The role of pathogens in diabetes pathogenesis and the potential of immunoproteomics as a diagnostic and prognostic tool. Front Microbiol 2022; 13:1042362. [PMID: 36483212 PMCID: PMC9724628 DOI: 10.3389/fmicb.2022.1042362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/26/2022] [Indexed: 09/11/2024] Open
Abstract
Diabetes mellitus (DM) is a group of metabolic diseases marked by hyperglycemia, which increases the risk of systemic infections. DM patients are at greater risk of hospitalization and mortality from bacterial, viral, and fungal infections. Poor glycemic control can result in skin, blood, bone, urinary, gastrointestinal, and respiratory tract infections and recurrent infections. Therefore, the evidence that infections play a critical role in DM progression and the hazard ratio for a person with DM dying from any infection is higher. Early diagnosis and better glycemic control can help prevent infections and improve treatment outcomes. Perhaps, half (49.7%) of the people living with DM are undiagnosed, resulting in a higher frequency of infections induced by the hyperglycemic milieu that favors immune dysfunction. Novel diagnostic and therapeutic markers for glycemic control and infection prevention are desirable. High-throughput blood-based immunoassays that screen infections and hyperglycemia are required to guide timely interventions and efficiently monitor treatment responses. The present review aims to collect information on the most common infections associated with DM, their origin, pathogenesis, and the potential of immunoproteomics assays in the early diagnosis of the infections. While infections are common in DM, their role in glycemic control and disease pathogenesis is poorly described. Nevertheless, more research is required to identify novel diagnostic and prognostic markers to understand DM pathogenesis and management of infections. Precise monitoring of diabetic infections by immunoproteomics may provide novel insights into disease pathogenesis and healthy prognosis.
Collapse
Affiliation(s)
| | | | - Andreas Oberbach
- Experimental Cardiac Surgery LMU Munich, Department of Cardiac Surgery, Ludwig Maximillian University of Munich, Munich, Germany
| | | | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine, Doha, Qatar
| |
Collapse
|
7
|
Masters EA, Ricciardi BF, Bentley KLDM, Moriarty TF, Schwarz EM, Muthukrishnan G. Skeletal infections: microbial pathogenesis, immunity and clinical management. Nat Rev Microbiol 2022; 20:385-400. [PMID: 35169289 PMCID: PMC8852989 DOI: 10.1038/s41579-022-00686-0] [Citation(s) in RCA: 274] [Impact Index Per Article: 91.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Osteomyelitis remains one of the greatest risks in orthopaedic surgery. Although many organisms are linked to skeletal infections, Staphylococcus aureus remains the most prevalent and devastating causative pathogen. Important discoveries have uncovered novel mechanisms of S. aureus pathogenesis and persistence within bone tissue, including implant-associated biofilms, abscesses and invasion of the osteocyte lacuno-canalicular network. However, little clinical progress has been made in the prevention and eradication of skeletal infection as treatment algorithms and outcomes have only incrementally changed over the past half century. In this Review, we discuss the mechanisms of persistence and immune evasion in S. aureus infection of the skeletal system as well as features of other osteomyelitis-causing pathogens in implant-associated and native bone infections. We also describe how the host fails to eradicate bacterial bone infections, and how this new information may lead to the development of novel interventions. Finally, we discuss the clinical management of skeletal infection, including osteomyelitis classification and strategies to treat skeletal infections with emerging technologies that could translate to the clinic in the future.
Collapse
Affiliation(s)
- Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin F Ricciardi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA.
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
8
|
The protease SplB of Staphylococcus aureus targets host complement components and inhibits complement-mediated bacterial opsonophagocytosis. J Bacteriol 2021; 204:e0018421. [PMID: 34633872 PMCID: PMC8765433 DOI: 10.1128/jb.00184-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that can cause life-threatening infections, particularly in immunocompromised individuals. The high-level virulence of S. aureus largely relies on its diverse and variable collection of virulence factors and immune evasion proteins, including the six serine protease-like proteins SplA to SplF. Spl proteins are expressed by most clinical isolates of S. aureus, but little is known about the molecular mechanisms by which these proteins modify the host’s immune response for the benefit of the bacteria. Here, we identify SplB as a protease that inactivates central human complement proteins, i.e., C3, C4, and the activation fragments C3b and C4b, by preferentially cleaving their α-chains. SplB maintained its proteolytic activity in human serum, degrading C3 and C4. SplB further cleaved the components of the terminal complement pathway, C5, C6, C7, C8, and C9. In contrast, the important soluble human complement regulators factor H and C4b-binding protein (C4BP), as well as C1q, were left intact. Thereby, SplB reduced C3b-mediated opsonophagocytosis by human neutrophils as well as C5b-9 deposition on the bacterial surface. In conclusion, we identified the first physiological substrates of the S. aureus extracellular protease SplB. This enzyme inhibits all three complement pathways and blocks opsonophagocytosis. Thus, SplB can be considered a novel staphylococcal complement evasion protein. IMPORTANCE The success of bacterial pathogens in immunocompetent humans depends on the control and inactivation of host immunity. S. aureus, like many other pathogens, efficiently blocks host complement attack early in infection. Aiming to understand the role of the S. aureus-encoded orphan proteases of the Spl operon, we asked whether these proteins play a role in immune escape. We found that SplB inhibits all three complement activation pathways as well as the lytic terminal complement pathway. This blocks the opsonophagocytosis of the bacteria by neutrophils. We also clarified the molecular mechanisms: SplB cleaves the human complement proteins C3, C4, C5, C6, C7, C8, and C9 as well as factor B but not the complement inhibitors factor H and C4BP. Thus, we identify the first physiological substrates of the extracellular protease SplB of S. aureus and characterize SplB as a novel staphylococcal complement evasion protein.
Collapse
|
9
|
Grace PS, Dolatshahi S, Lu LL, Cain A, Palmieri F, Petrone L, Fortune SM, Ottenhoff THM, Lauffenburger DA, Goletti D, Joosten SA, Alter G. Antibody Subclass and Glycosylation Shift Following Effective TB Treatment. Front Immunol 2021; 12:679973. [PMID: 34290702 PMCID: PMC8287567 DOI: 10.3389/fimmu.2021.679973] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
With an estimated 25% of the global population infected with Mycobacterium tuberculosis (Mtb), tuberculosis (TB) remains a leading cause of death by infectious diseases. Humoral immunity following TB treatment is largely uncharacterized, and antibody profiling could provide insights into disease resolution. Here we focused on the distinctive TB-specific serum antibody features in active TB disease (ATB) and compared them with latent TB infection (LTBI) or treated ATB (txATB). As expected, di-galactosylated glycan structures (lacking sialic acid) found on IgG-Fc differentiated LTBI from ATB, but also discriminated txATB from ATB. Moreover, TB-specific IgG4 emerged as a novel antibody feature that correlated with active disease, elevated in ATB, but significantly diminished after therapy. These findings highlight 2 novel TB-specific antibody changes that track with the resolution of TB and may provide key insights to guide TB therapy.
Collapse
Affiliation(s)
- Patricia S. Grace
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
- Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, MA, United States
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Lenette L. Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Adam Cain
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| | - Fabrizio Palmieri
- Clinical Department, National Institute for Infectious Diseases (INMI), IRCCS L. Spallanzani, Rome, Italy
| | - Linda Petrone
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS (INMI) L. Spallanzani, Rome, Italy
| | - Sarah M. Fortune
- Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, MA, United States
| | - Tom H. M. Ottenhoff
- Department of Infectious Disease, Leiden University Medical Center, Leiden, Netherlands
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Delia Goletti
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS (INMI) L. Spallanzani, Rome, Italy
| | - Simone A. Joosten
- Department of Infectious Disease, Leiden University Medical Center, Leiden, Netherlands
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| |
Collapse
|
10
|
Mrochen DM, Trübe P, Jorde I, Domanska G, van den Brandt C, Bröker BM. Immune Polarization Potential of the S. aureus Virulence Factors SplB and GlpQ and Modulation by Adjuvants. Front Immunol 2021; 12:642802. [PMID: 33936060 PMCID: PMC8081891 DOI: 10.3389/fimmu.2021.642802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Protection against Staphylococcus aureus is determined by the polarization of the anti-bacterial immune effector mechanisms. Virulence factors of S. aureus can modulate these and induce differently polarized immune responses in a single individual. We proposed that this may be due to intrinsic properties of the bacterial proteins. To test this idea, we selected two virulence factors, the serine protease-like protein B (SplB) and the glycerophosphoryl diester phosphodiesterase (GlpQ). In humans naturally exposed to S. aureus, SplB induces a type 2-biased adaptive immune response, whereas GlpQ elicits type 1/type 3 immunity. We injected the recombinant bacterial antigens into the peritoneum of S. aureus-naïve C57BL/6N mice and analyzed the immune response. This was skewed by SplB toward a Th2 profile including specific IgE, whereas GlpQ was weakly immunogenic. To elucidate the influence of adjuvants on the proteins’ polarization potential, we studied Montanide ISA 71 VG and Imject™Alum, which promote a Th1 and Th2 response, respectively. Alum strongly increased antibody production to the Th2-polarizing protein SplB, but did not affect the response to GlpQ. Montanide enhanced the antibody production to both S. aureus virulence factors. Montanide also augmented the inflammation in general, whereas Alum had little effect on the cellular immune response. The adjuvants did not override the polarization potential of the S. aureus proteins on the adaptive immune response.
Collapse
Affiliation(s)
- Daniel M Mrochen
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Patricia Trübe
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Ilka Jorde
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Grazyna Domanska
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Meyer TC, Michalik S, Holtfreter S, Weiss S, Friedrich N, Völzke H, Kocher T, Kohler C, Schmidt F, Bröker BM, Völker U. A Comprehensive View on the Human Antibody Repertoire Against Staphylococcus aureus Antigens in the General Population. Front Immunol 2021; 12:651619. [PMID: 33777051 PMCID: PMC7987813 DOI: 10.3389/fimmu.2021.651619] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Our goal was to provide a comprehensive overview of the antibody response to Staphylococcus aureus antigens in the general population as a basis for defining disease-specific profiles and diagnostic signatures. We tested the specific IgG and IgA responses to 79 staphylococcal antigens in 996 individuals from the population-based Study of Health in Pomerania. Using a dilution-based multiplex suspension array, we extended the dynamic range of specific antibody detection to seven orders of magnitude, allowing the precise quantification of high and low abundant antibody specificities in the same sample. The observed IgG and IgA antibody responses were highly heterogeneous with differences between individuals as well as between bacterial antigens that spanned several orders of magnitude. Some antigens elicited significantly more IgG than IgA and vice versa. We confirmed a strong influence of colonization on the antibody response and quantified the influence of sex, smoking, age, body mass index, and serum glucose on anti-staphylococcal IgG and IgA. However, all host parameters tested explain only a small part of the extensive variability in individual response to the different antigens of S. aureus.
Collapse
Affiliation(s)
- Tanja C Meyer
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Weiss
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Unit of Periodontology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Frank Schmidt
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,Proteomics Core, Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, Doha, Qatar
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Strom M, Crowley T, Shigdar S. Novel Detection of Nasty Bugs, Prevention Is Better than Cure. Int J Mol Sci 2020; 22:E149. [PMID: 33375709 PMCID: PMC7795740 DOI: 10.3390/ijms22010149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Hospital-acquired infections (HAIs) are a growing concern around the world. They contribute to increasing mortality and morbidity rates and are an economic threat. All hospital patients have the potential to contract an HAI, but those with weakened or inferior immune systems are at highest risk. Most hospital patients will contract at least one HAI, but many will contract multiple ones. Bacteria are the most common cause of HAIs and contribute to 80-90% of all HAIs, with Staphylococcus aureus, Clostridium difficile, Escherichia coli, Acinetobacter baumannii, Pseudomonas aeruginosa and Klebsiella pneumoniae accounting for the majority. Each of these bacteria are highly resistant to antibiotics and can produce a protective film, known as a biofilm, to further prevent their eradication. It has been shown that by detecting and eradicating bacteria in the environment, infection rates can be reduced. The current methods for detecting bacteria are time consuming, non-specific, and prone to false negatives or false positives. Aptamer-based biosensors have demonstrated specific, time-efficient and simple detection, highlighting the likelihood that they could be used in a similar way to detect HAI-causing bacteria.
Collapse
Affiliation(s)
- Mia Strom
- School of Medicine, Deakin University, Geelong 3216, Australia; (M.S.); (T.C.)
| | - Tamsyn Crowley
- School of Medicine, Deakin University, Geelong 3216, Australia; (M.S.); (T.C.)
- Centre for Molecular and Medical Research, Deakin University, Geelong 3216, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Geelong 3216, Australia; (M.S.); (T.C.)
- Centre for Molecular and Medical Research, Deakin University, Geelong 3216, Australia
| |
Collapse
|
13
|
Hu J, Gao M, Wang Z, Chen Y, Song Z, Xu H. Direct imaging of antigen-antibody binding by atomic force microscopy. APPLIED NANOSCIENCE 2020; 11:293-300. [PMID: 32989412 PMCID: PMC7511526 DOI: 10.1007/s13204-020-01558-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 09/15/2020] [Indexed: 11/06/2022]
Abstract
Direct observation of antigen–antibody binding at the nanoscale has always been a considerable challenging problem, and researchers have made tremendous efforts on it. In this study, the morphology of biotinylated antibody-specific Immunoglobulin E (IgE) immune complexes has been successfully imaged by atomic force microscopy (AFM) in the tapping-mode. The AFM images indicated that the individual immune complex was composed of an IgE and a biotinylated antibody. Excitingly, it is the first time that we have actually seen the IgE binding to biotinylated antibody. Alternatively, information on the length of IgE, biotinylated antibodies and biotinylated antibody-specific IgE immune complexes were also obtained, respectively. These results indicate the versatility of AFM technology in the identification of antigen–antibody binding. This work not only lays the basis for the direct imaging of the biotinylated antibody-IgE by AFM, but also offers valuable information for studying the targeted therapy and vaccine development in the future.
Collapse
Affiliation(s)
- Jing Hu
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China
| | - Mingyan Gao
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China
| | - Zuobin Wang
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China.,JR3CN and IRAC, University of Bedfordshire, Luton, LU1 3JU UK
| | - Yujuan Chen
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China.,School of Life Sciences, Changchun University of Science and Technology, Changchun, 130022 China
| | - Zhengxun Song
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China
| | - Hongmei Xu
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, 130022 China.,International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, 130022 China
| |
Collapse
|
14
|
Feuerstein R, Forde AJ, Lohrmann F, Kolter J, Ramirez NJ, Zimmermann J, Gomez de Agüero M, Henneke P. Resident macrophages acquire innate immune memory in staphylococcal skin infection. eLife 2020; 9:55602. [PMID: 32639232 PMCID: PMC7343389 DOI: 10.7554/elife.55602] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/26/2020] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a common colonizer of healthy skin and mucous membranes. At the same time, S. aureus is the most frequent cause of skin and soft tissue infections. Dermal macrophages (Mφ) are critical for the coordinated defense against invading S. aureus, yet they have a limited life span with replacement by bone marrow derived monocytes. It is currently poorly understood whether localized S. aureus skin infections persistently alter the resident Mφ subset composition and resistance to a subsequent infection. In a strictly dermal infection model we found that mice, which were previously infected with S. aureus, showed faster monocyte recruitment, increased bacterial killing and improved healing upon a secondary infection. However, skin infection decreased Mφ half-life, thereby limiting the duration of memory. In summary, resident dermal Mφ are programmed locally, independently of bone marrow-derived monocytes during staphylococcal skin infection leading to transiently increased resistance against a second infection.
Collapse
Affiliation(s)
- Reinhild Feuerstein
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron James Forde
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Florens Lohrmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Biology, University of Freiburg and IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Neftali Jose Ramirez
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jakob Zimmermann
- Maurice Müller Laboratories (Department for Biomedical Research), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | - Mercedes Gomez de Agüero
- Maurice Müller Laboratories (Department for Biomedical Research), Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Muthukrishnan G, Soin S, Beck CA, Grier A, Brodell JD, Lee CC, Ackert-Bicknell CL, Lee FEH, Schwarz EM, Daiss JL. A Bioinformatic Approach to Utilize a Patient's Antibody-Secreting Cells against Staphylococcus aureus to Detect Challenging Musculoskeletal Infections. Immunohorizons 2020; 4:339-351. [PMID: 32571786 DOI: 10.4049/immunohorizons.2000024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/04/2020] [Indexed: 01/22/2023] Open
Abstract
Noninvasive diagnostics for Staphylococcus aureus musculoskeletal infections (MSKI) remain challenging. Abs from newly activated, pathogen-specific plasmablasts in human blood, which emerge during an ongoing infection, can be used for diagnosing and tracking treatment response in diabetic foot infections. Using multianalyte immunoassays on medium enriched for newly synthesized Abs (MENSA) from Ab-secreting cells, we assessed anti-S. aureus IgG responses in 101 MSKI patients (63 culture-confirmed S. aureus, 38 S. aureus-negative) and 52 healthy controls. MENSA IgG levels were assessed for their ability to identify the presence and type of S. aureus MSKI using machine learning and multivariate receiver operating characteristic curves. Eleven S. aureus-infected patients were presented with prosthetic joint infections, 15 with fracture-related infections, 5 with native joint septic arthritis, 15 with diabetic foot infections, and 17 with suspected orthopedic infections in the soft tissue. Anti-S. aureus MENSA IgG levels in patients with non-S. aureus infections and healthy controls were 4-fold (***p = 0.0002) and 8-fold (****p < 0.0001) lower, respectively, compared with those with culture-confirmed S. aureus infections. Comparison of MENSA IgG responses among S. aureus culture-positive patients revealed Ags predictive of active MSKI (IsdB, SCIN, Gmd) and Ags predictive of MSKI type (IsdB, IsdH, Amd, Hla). When combined, IsdB, IsdH, Gmd, Amd, SCIN, and Hla were highly discriminatory of S. aureus MSKI (area under the ROC curve = 0.89 [95% confidence interval 0.82-0.93, p < 0.01]). Collectively, these results demonstrate the feasibility of a bioinformatic approach to use a patient's active immune proteome against S. aureus to diagnose challenging MSKI.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Sandeep Soin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Christopher A Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - James D Brodell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Charles C Lee
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| | - Cheryl L Ackert-Bicknell
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopedics, University of Colorado Denver, Denver, CO 80045; and
| | - Frances Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642.,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642.,Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642; .,Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
16
|
Kwiecinski JM, Horswill AR. Staphylococcus aureus bloodstream infections: pathogenesis and regulatory mechanisms. Curr Opin Microbiol 2020; 53:51-60. [PMID: 32172183 DOI: 10.1016/j.mib.2020.02.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus is an opportunistic pathogen that normally colonizes the human anterior nares. At the same time, this pathogen is one of the leading causes of life-threatening bloodstream infections, such as sepsis and endocarditis. In this review we will present the current understanding of the pathogenesis of these invasive infections, focusing on the mechanisms of S. aureus clearance from the bloodstream by the immune system, and how this pathogen hijacks the host defense and coagulation systems and further interacts with the blood vessel endothelium. Additionally, we will delve into the regulatory mechanisms S. aureus employs during an invasive infection. These new insights into host-pathogen interactions show promising avenues for the development of novel therapies for treating bloodstream infections.
Collapse
Affiliation(s)
- Jakub M Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, USA; Department of Veterans Affairs, Eastern Colorado Health Care System, Denver, USA.
| |
Collapse
|
17
|
Technical report: xMAPr – High-dynamic-range (HDR) quantification of antigen-specific antibody binding. J Proteomics 2020; 212:103577. [DOI: 10.1016/j.jprot.2019.103577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/28/2019] [Indexed: 11/19/2022]
|
18
|
Early-Stage Staphylococcus aureus Bloodstream Infection Causes Changes in the Concentrations of Lipoproteins and Acute-Phase Proteins and Is Associated with Low Antibody Titers against Bacterial Virulence Factors. mSystems 2020; 5:5/1/e00632-19. [PMID: 31964768 PMCID: PMC6977072 DOI: 10.1128/msystems.00632-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
S. aureus sepsis has a high complication and mortality rate. Given the limited therapeutic possibilities, effective prevention strategies, e.g., a vaccine, or the early identification of high-risk patients would be important but are not available. Our study showed an acute-phase response in patients with S. aureus bloodstream infection and evidence that lipoproteins are downregulated in plasma. Using immunoproteomics, stratification of patients appears to be achievable, since at the early stages of systemic S. aureus infection patients had low preexisting anti-S. aureus antibody levels. This strengthens the notion that a robust immune memory for S. aureus protects against infections with the pathogen. Systemic and quantitative investigations of human plasma proteins (proteomics) and Staphylococcus aureus-specific antibodies (immunoproteomics) provide complementary information and hold promise for the discovery of biomarkers in Staphylococcus aureus bloodstream infection (SABSI). Usually, data-dependent acquisition (DDA) is used for proteome analysis of serum or plasma, but data-independent acquisition (DIA) is more comprehensive and reproducible. In this prospective cohort study, we aimed to identify biomarkers associated with the early stages of SABSI using a serum DIA proteomic and immunoproteomic approach. Sera from 49 SABSI patients and 43 noninfected controls were analyzed. In total, 608 human serum proteins were identified with DIA. A total of 386 proteins could be quantified, of which 9 proteins, mainly belonging to acute-phase proteins, were significantly increased, while 7 high-density lipoproteins were lower in SABSI. In SABSI, total anti-S. aureus serum IgG was reduced compared with controls as shown by immunoproteomic quantification of IgG binding to 143 S. aureus antigens. IgG binding to 48 of these anti-S. aureus proteins was significantly lower in SABSI, while anti-Ecb IgG was the only one increased in SABSI. Serum IgG binding to autoinducing peptide MsrB, FadB, EsxA, Pbp2, FadB, SspB, or SodA was very low in SABSI. This marker panel discriminated early SABSI from controls with 95% sensitivity and 100% specificity according to random forest prediction. This holds promise for patient stratification according to their risk of S. aureus infection, underlines the protective function of the adaptive immune system, and encourages further efforts in the development of a vaccine against S. aureus. IMPORTANCES. aureus sepsis has a high complication and mortality rate. Given the limited therapeutic possibilities, effective prevention strategies, e.g., a vaccine, or the early identification of high-risk patients would be important but are not available. Our study showed an acute-phase response in patients with S. aureus bloodstream infection and evidence that lipoproteins are downregulated in plasma. Using immunoproteomics, stratification of patients appears to be achievable, since at the early stages of systemic S. aureus infection patients had low preexisting anti-S. aureus antibody levels. This strengthens the notion that a robust immune memory for S. aureus protects against infections with the pathogen.
Collapse
|
19
|
Muthukrishnan G, Masters EA, Daiss JL, Schwarz EM. Mechanisms of Immune Evasion and Bone Tissue Colonization That Make Staphylococcus aureus the Primary Pathogen in Osteomyelitis. Curr Osteoporos Rep 2019; 17:395-404. [PMID: 31721069 PMCID: PMC7344867 DOI: 10.1007/s11914-019-00548-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Staphylococcus aureus is the primary pathogen responsible for osteomyelitis, which remains a major healthcare burden. To understand its dominance, here we review the unique pathogenic mechanisms utilized by S. aureus that enable it to cause incurable osteomyelitis. RECENT FINDINGS Using an arsenal of toxins and virulence proteins, S. aureus kills and usurps immune cells during infection, to produce non-neutralizing pathogenic antibodies that thwart adaptive immunity. S. aureus also has specific mechanisms for distinct biofilm formation on implants, necrotic bone tissue, bone marrow, and within the osteocyte lacuno-canicular networks (OLCN) of live bone. In vitro studies have also demonstrated potential for intracellular colonization of osteocytes, osteoblasts, and osteoclasts. S. aureus has evolved a multitude of virulence mechanisms to achieve life-long infection of the bone, most notably colonization of OLCN. Targeting S. aureus proteins involved in these pathways could provide new targets for antibiotics and immunotherapies.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
20
|
Teufelberger AR, Bröker BM, Krysko DV, Bachert C, Krysko O. Staphylococcus aureus Orchestrates Type 2 Airway Diseases. Trends Mol Med 2019; 25:696-707. [PMID: 31176612 DOI: 10.1016/j.molmed.2019.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022]
Abstract
Staphylococcus aureus persistently colonizes the nostrils of one-third of the population but colonizes the sinus mucosa in up to 90% of patients with nasal polyps, implying a possible role in airway disease. Recent findings give new mechanistic insights into the ability of S. aureus to trigger type 2 inflammatory responses in the upper and lower airways. This novel concept of a S. aureus-driven chronic airway inflammatory disease suggests a new understanding of disease triggers. This article reviews the role of S. aureus in chronic inflammatory airway diseases and discusses possible therapeutic approaches to target S. aureus.
Collapse
Affiliation(s)
- Andrea R Teufelberger
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Dmitri V Krysko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia; Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium; Division of ENT Diseases, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Olga Krysko
- Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium.
| |
Collapse
|
21
|
Scheuch M, Freiin von Rheinbaben S, Kabisch A, Engeßer J, Ahrendt S, Dabers T, Kohler C, Holtfreter S, Bröker BM, Stracke S. Staphylococcus aureus colonization in hemodialysis patients: a prospective 25 months observational study. BMC Nephrol 2019; 20:153. [PMID: 31060511 PMCID: PMC6503363 DOI: 10.1186/s12882-019-1332-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/12/2019] [Indexed: 12/24/2022] Open
Abstract
Background Dialysis patients are frequently exposed to Staphylococcus aureus due to stays in dialysis centers, hospitals or rest homes. The hemodialysis vascular access is a potential entry site for S. aureus, in particular when using a central venous catheter (CVC) which increases the risk of sepsis compared to arteriovenous (AV) fistula. We prospectively followed a cohort of 86 hemodialysis patients from an outpatient dialysis center over 25 months analyzing S. aureus carrier status, S. aureus infection rates and mortality. Methods Demographic data and patients´ medical histories were collected and followed from all hemodialysis patients. Blood samples, nasal swabs and swabs from the hemodialysis vascular access site were taken every six months for a period of 25 months and tested for S. aureus. Strains were cultured and further characterized by spa PCR and microarray-based genotyping. Resulting data were compared with those from the general population. Results In cross-sectional analyses, an average of 40% of hemodialysis patients were S. aureus carriers compared to 27% in the general population. Longitudinally, a total of 65% were S. aureus carriers: 16% were persistent carriers, 43% were intermittently colonized. The most common S. aureus lineage in the dialysis patient cohort was the clonal complex (CC) 8 and the spa type t008, while in the general population, the clonal complex CC30 dominates. During the study period, we observed six S. aureus-associated blood stream infections with one S. aureus attributable death. S. aureus carriers with an AV fistula were more densely colonized in the nasal mucosa compared to patients with a CVC. Overall mortality was lower for hemodialysis patients with a positive S. aureus carrier status compared to non-carriers (hazard ratio of 0.19). Conclusions Compared to the general population, hemodialysis patients were more frequently colonized with S. aureus and displayed both different S. aureus colonization densities as well as lineages, possibly explained by more frequent exposure to health care environments. The lower overall mortality in carriers compared to non-carriers is intriguing and will be investigated in detail in the future. Trial registration ISRCTN 14385893, 2. October 2018, retrospectively registered. Electronic supplementary material The online version of this article (10.1186/s12882-019-1332-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthias Scheuch
- Department of Internal Medicine A, Division of Nephrology, University Medicine Greifswald, Greifswald, Germany
| | | | - Antje Kabisch
- Department of Internal Medicine A, Division of Nephrology, University Medicine Greifswald, Greifswald, Germany
| | - Jonas Engeßer
- Department of Internal Medicine A, Division of Nephrology, University Medicine Greifswald, Greifswald, Germany
| | - Susanne Ahrendt
- Kuratorium für Dialyse und Nierentransplantation e.V., KfH-Nierenzentrum Greifswald, Greifswald, Germany
| | - Thomas Dabers
- Department of Internal Medicine A, Division of Nephrology, University Medicine Greifswald, Greifswald, Germany.,Kuratorium für Dialyse und Nierentransplantation e.V., KfH-Nierenzentrum Greifswald, Greifswald, Germany
| | - Christian Kohler
- Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Sylvia Stracke
- Department of Internal Medicine A, Division of Nephrology, University Medicine Greifswald, Greifswald, Germany. .,Kuratorium für Dialyse und Nierentransplantation e.V., KfH-Nierenzentrum Greifswald, Greifswald, Germany.
| |
Collapse
|
22
|
Kodani M, Martin M, de Castro VL, Drobeniuc J, Kamili S. An Automated Immunoblot Method for Detection of IgG Antibodies to Hepatitis C Virus: a Potential Supplemental Antibody Confirmatory Assay. J Clin Microbiol 2019; 57:e01567-18. [PMID: 30651390 PMCID: PMC6425170 DOI: 10.1128/jcm.01567-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/03/2019] [Indexed: 01/16/2023] Open
Abstract
An estimated 41,200 people were newly infected with hepatitis C virus (HCV) in 2016 in the United States. Screening tests for antibodies to HCV may generate up to 32% false positivity in low-risk populations. Current Centers for Disease Control and Prevention (CDC) screening recommendations do not require confirmatory testing of a screening anti-HCV-positive test; however, confirmation is valuable for surveillance in the absence of HCV RNA testing. A recombinant immunoblot assay (RIBA) was used as a confirmatory assay for anti-HCV-reactive samples but was discontinued in 2013. Another anti-HCV confirmatory assay, INNO-LIA, is commercially available in Europe but is not approved by the Food and Drug Administration (FDA) in the United States. We report the development of an anti-HCV assay that was performed on an automated immunoblot platform using a fourth-generation HCV recombinant fusion protein. Based on testing of 70 well-characterized samples, of which 40 were HCV RNA and anti-HCV positive, 15 were HCV RNA positive/anti-HCV negative, and 15 were HCV RNA and anti-HCV negative, the specificity and sensitivity of the HCV-WES assay were 100% and 95%, respectively. Concordance between INNO-LIA and HCV-WES was determined by testing 205 HCV RNA-negative/anti-HCV-positive samples, of which 149 (72.7%) were positive by HCV-WES, while 146 (71.2%) were positive by INNO-LIA. We have shown proof of concept for the use of this test for confirmation of screened anti-HCV results. The HCV-WES assay has advantages over manual Western blot assays and INNO-LIA, including ease of use, lower cost, and reduced hands-on time.
Collapse
Affiliation(s)
- Maja Kodani
- Division of Viral Hepatitis, National Center for HIV, Hepatitis, STD and Tuberculosis Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Miranda Martin
- Division of Viral Hepatitis, National Center for HIV, Hepatitis, STD and Tuberculosis Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Vivianne Landgraf de Castro
- Division of Viral Hepatitis, National Center for HIV, Hepatitis, STD and Tuberculosis Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jan Drobeniuc
- Division of Viral Hepatitis, National Center for HIV, Hepatitis, STD and Tuberculosis Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Saleem Kamili
- Division of Viral Hepatitis, National Center for HIV, Hepatitis, STD and Tuberculosis Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
23
|
Rigat F, Bartolini E, Dalsass M, Kumar N, Marchi S, Speziale P, Maione D, Chen L, Romano MR, Alegre ML, Bagnoli F, Daum RS, David MZ. Retrospective Identification of a Broad IgG Repertoire Differentiating Patients With S. aureus Skin and Soft Tissue Infections From Controls. Front Immunol 2019; 10:114. [PMID: 30792711 PMCID: PMC6375365 DOI: 10.3389/fimmu.2019.00114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Although the relevance of humoral immunity for protection against S. aureus skin and soft tissue infections (SSTIs) has been suggested by several animal and human studies, the question of which human antibodies may be protective has so far impeded the development of a safe and effective vaccine. Because most adults have developed certain anti-S. aureus antibodies due to S. aureus colonization or infection, we hypothesized that the titers of antibodies to S. aureus in uninfected controls would differ from those in infected patients and would also differ in infected patients from the time of acute infection to a 40-day convalescent serum. Methods: To test these hypotheses, we measured human antibody levels against a panel of 134 unique antigens comprising the S. aureus surfome and secretome in subjects with active culture-confirmed S. aureus SSTIs (cases) and in controls with no infection, using a novel S. aureus protein microarray. Results: Most S. aureus SSTI patients (n = 60) and controls (n = 142) had antibodies to many of the tested S. aureus antigens. Univariate analysis showed statistically weak differences in the IgG levels to some antigens in the SSTI patient (case) sera compared with controls. Antibody levels to most tested antigens did not increase comparing acute with 40-day serum. Multiple logistic regression identified a rich subset of antigens that, by their antibody levels, together correctly differentiated all cases from all controls. Conclusions: Antibodies directed against S. aureus antigens were present both in patients with S. aureus SSTIs and in uninfected control patients. We found that SSTI patients and controls could be distinguished only based on differences in antibody levels to many staphylococcal surface and secreted antigens. Our results demonstrate that in the studied population, the levels of anti-S. aureus antibodies appear largely fixed, suggesting that there may be some level of unresponsiveness to natural infection.
Collapse
Affiliation(s)
- Fabio Rigat
- GSK Pharmaceuticals R&D, Stevenage, United Kingdom
| | | | | | - Neha Kumar
- Department of Pediatrics, University of Chicago, Chicago, IL, United States
| | | | - Pietro Speziale
- Department of Engineering, University of Pavia, Pavia, Italy
- Biochemistry Section, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Luqiu Chen
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | | | - Maria-Luisa Alegre
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | | | - Robert S. Daum
- Department of Pediatrics, University of Chicago, Chicago, IL, United States
| | - Michael Z. David
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
Raafat D, Otto M, Reppschläger K, Iqbal J, Holtfreter S. Fighting Staphylococcus aureus Biofilms with Monoclonal Antibodies. Trends Microbiol 2019; 27:303-322. [PMID: 30665698 DOI: 10.1016/j.tim.2018.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen and one of the most frequent causes of biofilm-related infections. The treatment of S. aureus biofilms is hampered by the ability of the biofilm structure to shield bacteria from antibiotics as well as the host's immune system. Therefore, new preventive and/or therapeutic interventions, including the use of antibody-based approaches, are urgently required. In this review, we describe the mechanisms by which anti-S. aureus antibodies can help in combating biofilms, including an up-to-date overview of monoclonal antibodies currently in clinical trials. Moreover, we highlight ongoing efforts in passive vaccination against S. aureus biofilm infections, with special emphasis on promising targets, and finally indicate the direction into which future research could be heading.
Collapse
Affiliation(s)
- Dina Raafat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Egypt; Current affiliation: Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Kevin Reppschläger
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jawad Iqbal
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
25
|
Flaxman A, Yamaguchi Y, van Diemen PM, Rollier C, Allen E, Elshina E, Wyllie DH. Heterogeneous early immune responses to the S. aureus EapH2 antigen induced by gastrointestinal tract colonisation impact the response to subsequent vaccination. Vaccine 2019; 37:494-501. [PMID: 30503080 DOI: 10.1016/j.vaccine.2018.11.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022]
Abstract
INTRODUCTION S. aureus is a pathogen to which individuals are exposed shortly after birth, with immune responses to S. aureus increasing during childhood. There is marked heterogeneity between the anti- S. aureus immune responses of different humans, the basis of which is not fully understood. METHODS To investigate development of anti-S. aureus immune responses, we studied S. aureus colonised mice under controlled conditions. Mice were either acquired colonised from breeding colonies, or experimentally colonised by exposure to a cage environment which had been sprayed with a S. aureus suspension. Colonisation was monitored by sequential stool sampling, and immunoglobulin levels against both whole fixed S. aureus and individual S. aureus antigens quantified. The immunological impact of colonisation on subsequent vaccination was investigated. RESULTS Colonised BALB/c and BL/6 mice develop serum anti- S. aureus cell surface IgG1 antibodies. Responses were proportional to the cumulative S. aureus bioburden in the mice, and were higher in BALB/c mice, which have higher colonisation levels, than in C57BL/6 animals. We observed marked variation in the induction of anti-cell surface antibodies, even in genetically identical mice experimentally colonised with the same S. aureus clone. Heterogeneity was also evident when monitoring immune responses to the secreted S. aureus protein EapH2. Approximately 50% of colonised mice developed anti-EapH2 responses (responders); in other mice, responses were not significantly different to those in uncolonised mice (non-responders). Following vaccination with a replication deficient adenovirus expressing EapH2, less anti-EapH2 antibody was generated in non-responder than responder animals. CONCLUSIONS In genetically identical mice, S. aureus colonisation results in all-or-nothing antibody responses against some antigens, including EapH2. For antigens involved in colonisation success by microbes, apparently stochastic early immune responses may impact both vaccine responses and the establishment of an animal-specific microbiome.
Collapse
Affiliation(s)
- Amy Flaxman
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Yuko Yamaguchi
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Pauline M van Diemen
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Christine Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Biomedical Research Centre, CCVTM, Churchill Drive, UK
| | - Elizabeth Allen
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Elizaveta Elshina
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - David H Wyllie
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK.
| |
Collapse
|
26
|
Gerlach D, Guo Y, De Castro C, Kim SH, Schlatterer K, Xu FF, Pereira C, Seeberger PH, Ali S, Codée J, Sirisarn W, Schulte B, Wolz C, Larsen J, Molinaro A, Lee BL, Xia G, Stehle T, Peschel A. Methicillin-resistant Staphylococcus aureus alters cell wall glycosylation to evade immunity. Nature 2018; 563:705-709. [PMID: 30464342 DOI: 10.1038/s41586-018-0730-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/18/2018] [Indexed: 01/19/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a frequent cause of difficult-to-treat, often fatal infections in humans1,2. Most humans have antibodies against S. aureus, but these are highly variable and often not protective in immunocompromised patients3. Previous vaccine development programs have not been successful4. A large percentage of human antibodies against S. aureus target wall teichoic acid (WTA), a ribitol-phosphate (RboP) surface polymer modified with N-acetylglucosamine (GlcNAc)5,6. It is currently unknown whether the immune evasion capacities of MRSA are due to variation of dominant surface epitopes such as those associated with WTA. Here we show that a considerable proportion of the prominent healthcare-associated and livestock-associated MRSA clones CC5 and CC398, respectively, contain prophages that encode an alternative WTA glycosyltransferase. This enzyme, TarP, transfers GlcNAc to a different hydroxyl group of the WTA RboP than the standard enzyme TarS7, with important consequences for immune recognition. TarP-glycosylated WTA elicits 7.5-40-fold lower levels of immunoglobulin G in mice than TarS-modified WTA. Consistent with this, human sera contained only low levels of antibodies against TarP-modified WTA. Notably, mice immunized with TarS-modified WTA were not protected against infection with tarP-expressing MRSA, indicating that TarP is crucial for the capacity of S. aureus to evade host defences. High-resolution structural analyses of TarP bound to WTA components and uridine diphosphate GlcNAc (UDP-GlcNAc) explain the mechanism of altered RboP glycosylation and form a template for targeted inhibition of TarP. Our study reveals an immune evasion strategy of S. aureus based on averting the immunogenicity of its dominant glycoantigen WTA. These results will help with the identification of invariant S. aureus vaccine antigens and may enable the development of TarP inhibitors as a new strategy for rendering MRSA susceptible to human host defences.
Collapse
Affiliation(s)
- David Gerlach
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Yinglan Guo
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Naples, Naples, Italy
| | - Sun-Hwa Kim
- National Research Laboratory of Defense Proteins, College of Pharmacy, Pusan National University, Pusan, South Korea
| | - Katja Schlatterer
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Fei-Fei Xu
- Max-Planck-Institute for Colloids and Interfaces, Potsdam, Germany
| | - Claney Pereira
- Max-Planck-Institute for Colloids and Interfaces, Potsdam, Germany
| | | | - Sara Ali
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Wanchat Sirisarn
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Berit Schulte
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Interfaculty Institute of Microbiology and Infection Medicine, Medical Microbiology, University of Tübingen, Tübingen, Germany
| | - Christiane Wolz
- German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Interfaculty Institute of Microbiology and Infection Medicine, Medical Microbiology, University of Tübingen, Tübingen, Germany
| | - Jesper Larsen
- Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples, Naples, Italy
| | - Bok Luel Lee
- National Research Laboratory of Defense Proteins, College of Pharmacy, Pusan National University, Pusan, South Korea
| | - Guoqing Xia
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany. .,Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
27
|
Darisipudi MN, Nordengrün M, Bröker BM, Péton V. Messing with the Sentinels-The Interaction of Staphylococcus aureus with Dendritic Cells. Microorganisms 2018; 6:microorganisms6030087. [PMID: 30111706 PMCID: PMC6163568 DOI: 10.3390/microorganisms6030087] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/12/2018] [Accepted: 08/14/2018] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a dangerous pathogen as well as a frequent colonizer, threatening human health worldwide. Protection against S. aureus infection is challenging, as the bacteria have sophisticated strategies to escape the host immune response. To maintain equilibrium with S. aureus, both innate and adaptive immune effector mechanisms are required. Dendritic cells (DCs) are critical players at the interface between the two arms of the immune system, indispensable for inducing specific T cell responses. In this review, we highlight the importance of DCs in mounting innate as well as adaptive immune responses against S. aureus with emphasis on their role in S. aureus-induced respiratory diseases. We also review what is known about mechanisms that S. aureus has adopted to evade DCs or manipulate these cells to its advantage.
Collapse
Affiliation(s)
- Murthy N Darisipudi
- Department of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße DZ7, D-17475 Greifswald, Germany.
| | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße DZ7, D-17475 Greifswald, Germany.
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße DZ7, D-17475 Greifswald, Germany.
| | - Vincent Péton
- Department of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße DZ7, D-17475 Greifswald, Germany.
| |
Collapse
|
28
|
Jorge AM, Schneider J, Unsleber S, Xia G, Mayer C, Peschel A. Staphylococcus aureus counters phosphate limitation by scavenging wall teichoic acids from other staphylococci via the teichoicase GlpQ. J Biol Chem 2018; 293:14916-14924. [PMID: 30068554 DOI: 10.1074/jbc.ra118.004584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/26/2018] [Indexed: 11/06/2022] Open
Abstract
Staphylococcus aureus is part of the human nasal and skin microbiomes along with other bacterial commensals and opportunistic pathogens. Nutrients are scarce in these habitats, demanding effective nutrient acquisition and competition strategies. How S. aureus copes with phosphate limitation is still unknown. Wall teichoic acid (WTA), a polyol-phosphate polymer, could serve as a phosphate source, but whether S. aureus can utilize it during phosphate starvation remains unknown. S. aureus secretes a glycerophosphodiesterase, GlpQ, that cleaves a broad variety of glycerol-3-phosphate (GroP) headgroups of deacylated phospholipids, providing this bacterium with GroP as a carbon and phosphate source. Here we demonstrate that GlpQ can also use glycerophosphoglycerol derived from GroP WTA from coagulase-negative Staphylococcus lugdunensis, Staphylococcus capitis, and Staphylococcus epidermidis, which share the nasal and skin habitats with S. aureus Therefore, S. aureus GlpQ is the first reported WTA-hydrolyzing enzyme, or teichoicase, from Staphylococcus Activity assays revealed that unmodified WTA is the preferred GlpQ substrate, and the results from MS analysis suggested that GlpQ uses an exolytic cleavage mechanism. Importantly, GlpQ did not hydrolyze the ribitol-5-phosphate WTA polymers of S. aureus, underscoring its role in interspecies competition rather than in S. aureus cell wall homeostasis or WTA recycling. glpQ expression was strongly up-regulated under phosphate limitation, and GlpQ allowed S. aureus to grow in the presence of GroP WTA as the sole phosphate source. Our study reveals a novel and unprecedented strategy of S. aureus for acquiring phosphate from bacterial competitors under the phosphate-limiting conditions in the nasal or skin environments.
Collapse
Affiliation(s)
- Ana Maria Jorge
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany, .,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Jonathan Schneider
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Sandra Unsleber
- the Microbiology/Biotechnology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
| | - Guoqing Xia
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Christoph Mayer
- the Microbiology/Biotechnology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
| | - Andreas Peschel
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| |
Collapse
|
29
|
From the genome sequence via the proteome to cell physiology – Pathoproteomics and pathophysiology of Staphylococcus aureus. Int J Med Microbiol 2018; 308:545-557. [DOI: 10.1016/j.ijmm.2018.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/23/2017] [Accepted: 01/02/2018] [Indexed: 02/01/2023] Open
|
30
|
Lee AS, de Lencastre H, Garau J, Kluytmans J, Malhotra-Kumar S, Peschel A, Harbarth S. Methicillin-resistant Staphylococcus aureus. Nat Rev Dis Primers 2018; 4:18033. [PMID: 29849094 DOI: 10.1038/nrdp.2018.33] [Citation(s) in RCA: 856] [Impact Index Per Article: 122.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the 1960s, methicillin-resistant Staphylococcus aureus (MRSA) has emerged, disseminated globally and become a leading cause of bacterial infections in both health-care and community settings. However, there is marked geographical variation in MRSA burden owing to several factors, including differences in local infection control practices and pathogen-specific characteristics of the circulating clones. Different MRSA clones have resulted from the independent acquisition of staphylococcal cassette chromosome mec (SCCmec), which contains genes encoding proteins that render the bacterium resistant to most β-lactam antibiotics (such as methicillin), by several S. aureus clones. The success of MRSA is a consequence of the extensive arsenal of virulence factors produced by S. aureus combined with β-lactam resistance and, for most clones, resistance to other antibiotic classes. Clinical manifestations of MRSA range from asymptomatic colonization of the nasal mucosa to mild skin and soft tissue infections to fulminant invasive disease with high mortality. Although treatment options for MRSA are limited, several new antimicrobials are under development. An understanding of colonization dynamics, routes of transmission, risk factors for progression to infection and conditions that promote the emergence of resistance will enable optimization of strategies to effectively control MRSA. Vaccine candidates are also under development and could become an effective prevention measure.
Collapse
Affiliation(s)
- Andie S Lee
- Departments of Infectious Diseases and Microbiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Hermínia de Lencastre
- Laboratory of Microbiology and Infectious Diseases, The Rockefeller University, New York, NY, USA.,Laboratory of Molecular Genetics, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Javier Garau
- Department of Medicine, Hospital Universitari Mutua de Terrassa, Barcelona, Spain
| | - Jan Kluytmans
- Department of Infection Control, Amphia Hospital, Breda, Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Universiteit Antwerpen, Wilrijk, Belgium
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Stephan Harbarth
- Infection Control Programme, University of Geneva Hospitals and Faculty of Medicine, WHO Collaborating Center, Geneva, Switzerland
| |
Collapse
|
31
|
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes superficial and invasive infections in the hospital and community. High mortality from infection emphasizes the need for improved methods for prevention and treatment. Although S. aureus possesses an arsenal of virulence factors that contribute to evasion of host defenses, few studies have examined long-term humoral and B-cell responses. Adults with acute-phase skin and soft tissue infections were recruited; blood samples were obtained; and S. aureus isolates, including methicillin-resistant strains, were subjected to genomic sequence analysis. In comparisons of acute-phase sera with convalescent-phase sera, a minority (37.5%) of patients displayed 2-fold or greater increases in antibody titers against three or more S. aureus antigens, whereas nearly half exhibited no changes, despite the presence of toxin genes in most infecting strains. Moreover, enhanced antibody responses waned over time, which could reflect a defect in B-cell memory or long-lived plasma cells. However, memory B cells reactive with a range of S. aureus antigens were prevalent at both acute-phase and convalescent-phase time points. While some memory B cells exhibited toxin-specific binding, those cross-reactive with structurally related leucocidin subunits were dominant across patients, suggesting the targeting of conserved epitopes. Memory B-cell reactivity correlated with serum antibody levels for selected S. aureus exotoxins, suggesting a relationship between the cellular and humoral compartments. Overall, although there was no global defect in the representation of anti-S. aureus memory B cells, there was evidence of restrictions in the range of epitopes recognized, which may suggest potential therapeutic approaches for augmenting host defenses. The contribution of B-cell memory and long-term antibody responses to host defenses against S. aureus exotoxins remains poorly understood. Our studies confirmed that infection did not commonly lead to enhanced long-term humoral responses. Whereas circulating memory B cells against S. aureus secreted exotoxins were prevalent, they were dominated by cross-reactivity with structurally related leucocidin subunits, consistent with recognition of conserved epitopes. These findings also provide the first evidence of a relationship between the reactivity of antistaphylococcal circulating memory B cells and serum antibody levels. In general, infection was not associated with a global defect in B-cell memory for S. aureus secreted factors, and responses were highly dominated by cross-reactivity to structurally related exotoxins, which arguably may alone be suboptimal in providing host defenses. Our studies illuminate aspects of the S. aureus-host relationship that may better inform strategies for the development of an effective protective vaccine.
Collapse
|
32
|
Mrochen DM, Grumann D, Schulz D, Gumz J, Trübe P, Pritchett-Corning K, Johnson S, Nicklas W, Kirsch P, Martelet K, Brandt JVD, Berg S, Bröker BM, Wiles S, Holtfreter S. Global spread of mouse-adapted Staphylococcus aureus lineages CC1, CC15, and CC88 among mouse breeding facilities. Int J Med Microbiol 2017; 308:598-606. [PMID: 29174495 DOI: 10.1016/j.ijmm.2017.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/27/2017] [Accepted: 11/16/2017] [Indexed: 11/25/2022] Open
Abstract
We previously reported that laboratory mice from all global vendors are frequently colonized with Staphylococcus aureus (S. aureus). Genotyping of a snap sample of murine S. aureus isolates from Charles River, US, showed that mice were predominantly colonized with methicillin-sensitive CC88 strains. Here, we expanded our view and investigated whether laboratory mice from other global animal facilities are colonized with similar strains or novel S. aureus lineages, and whether the murine S. aureus isolates show features of host adaptation. In total, we genotyped 230 S. aureus isolates from various vendor facilities of laboratory mice around the globe (Charles River facilities in the USA, Canada, France, and Germany; another US facility) and university- or company-associated breeding facilities in Germany, China and New Zealand. Spa typing was performed to analyse the clonal relationship of the isolates. Moreover, multiplex PCRs were performed for human-specific virulence factors, the immune-evasion cluster (IEC) and superantigen genes (SAg). We found a total of 58 different spa types that clustered into 15 clonal complexes (CCs). Three of these S. aureus lineages had spread globally among laboratory mice and accounted for three quarters of the isolates: CC1 (13.5%), CC15 (14.3%), and CC88 (47.0%). Compared to human colonizing isolates of the same lineages, the murine isolates frequently lacked IEC genes and SAg genes on mobile genetic elements, implying long-term adaptation to the murine host. In conclusion, laboratory mice from various vendors are colonized with host-adapted S. aureus-strains of a few lineages, predominantly the CC88 lineage. S. aureus researchers must be cautioned that S. aureus colonization might be a relevant confounder in infection and vaccination studies and are therefore advised to screen their mice before experimentation.
Collapse
Affiliation(s)
- Daniel M Mrochen
- Department of Immunology, University Medicine Greifswald, Germany
| | - Dorothee Grumann
- Department of Immunology, University Medicine Greifswald, Germany
| | - Daniel Schulz
- Department of Immunology, University Medicine Greifswald, Germany
| | - Janine Gumz
- Department of Immunology, University Medicine Greifswald, Germany
| | - Patricia Trübe
- Department of Immunology, University Medicine Greifswald, Germany
| | | | - Sarah Johnson
- Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | - Werner Nicklas
- Deutsches Krebsforschungszentrum, Microbiological Diagnostics, Heidelberg, Germany
| | - Petra Kirsch
- Tierforschungszentrum, University of Ulm, Ulm, Germany
| | - Karine Martelet
- Charles River, 360 diagnostic (RADS France), L'Arbresle cedex, France
| | - Jens van den Brandt
- Central Core & Research Facility of Laboratory Animals, University Medicine Greifswald, Germany
| | - Sabine Berg
- Central Core & Research Facility of Laboratory Animals, University Medicine Greifswald, Germany
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Germany
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Germany.
| |
Collapse
|
33
|
Schulz D, Grumann D, Trübe P, Pritchett-Corning K, Johnson S, Reppschläger K, Gumz J, Sundaramoorthy N, Michalik S, Berg S, van den Brandt J, Fister R, Monecke S, Uy B, Schmidt F, Bröker BM, Wiles S, Holtfreter S. Laboratory Mice Are Frequently Colonized with Staphylococcus aureus and Mount a Systemic Immune Response-Note of Caution for In vivo Infection Experiments. Front Cell Infect Microbiol 2017; 7:152. [PMID: 28512627 PMCID: PMC5411432 DOI: 10.3389/fcimb.2017.00152] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/11/2017] [Indexed: 11/16/2022] Open
Abstract
Whether mice are an appropriate model for S. aureus infection and vaccination studies is a matter of debate, because they are not considered as natural hosts of S. aureus. We previously identified a mouse-adapted S. aureus strain, which caused infections in laboratory mice. This raised the question whether laboratory mice are commonly colonized with S. aureus and whether this might impact on infection experiments. Publicly available health reports from commercial vendors revealed that S. aureus colonization is rather frequent, with rates as high as 21% among specific-pathogen-free mice. In animal facilities, S. aureus was readily transmitted from parents to offspring, which became persistently colonized. Among 99 murine S. aureus isolates from Charles River Laboratories half belonged to the lineage CC88 (54.5%), followed by CC15, CC5, CC188, and CC8. A comparison of human and murine S. aureus isolates revealed features of host adaptation. In detail, murine strains lacked hlb-converting phages and superantigen-encoding mobile genetic elements, and were frequently ampicillin-sensitive. Moreover, murine CC88 isolates coagulated mouse plasma faster than human CC88 isolates. Importantly, S. aureus colonization clearly primed the murine immune system, inducing a systemic IgG response specific for numerous S. aureus proteins, including several vaccine candidates. Phospholipase C emerged as a promising test antigen for monitoring S. aureus colonization in laboratory mice. In conclusion, laboratory mice are natural hosts of S. aureus and therefore, could provide better infection models than previously assumed. Pre-exposure to the bacteria is a possible confounder in S. aureus infection and vaccination studies and should be monitored.
Collapse
Affiliation(s)
- Daniel Schulz
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | - Dorothee Grumann
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | - Patricia Trübe
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | | | - Sarah Johnson
- Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of AucklandAuckland, New Zealand
| | - Kevin Reppschläger
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | - Janine Gumz
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | - Nandakumar Sundaramoorthy
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, ZIK FunGene, University Medicine GreifswaldGreifswald, Germany
| | - Stephan Michalik
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, ZIK FunGene, University Medicine GreifswaldGreifswald, Germany
| | - Sabine Berg
- Central Core and Research Facility of Laboratory Animals, University Medicine GreifswaldGreifswald, Germany
| | - Jens van den Brandt
- Central Core and Research Facility of Laboratory Animals, University Medicine GreifswaldGreifswald, Germany
| | - Richard Fister
- Charles River, Research and Professional ServicesWilmington, MA, USA
| | - Stefan Monecke
- Alere TechnologiesJena, Germany.,Institute for Medical Microbiology and Hygiene, Medical Faculty "Carl Gustav Carus"Dresden, Germany
| | - Benedict Uy
- Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of AucklandAuckland, New Zealand
| | - Frank Schmidt
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, ZIK FunGene, University Medicine GreifswaldGreifswald, Germany
| | - Barbara M Bröker
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| | - Siouxsie Wiles
- Bioluminescent Superbugs Lab, Department of Molecular Medicine and Pathology, University of AucklandAuckland, New Zealand.,Maurice Wilkins Centre for Molecular BiodiscoveryAuckland, New Zealand
| | - Silva Holtfreter
- Department of Immunology, University Medicine GreifswaldGreifswald, Germany
| |
Collapse
|
34
|
Jiang L, Ren H, Zhou H, Qin T, Chen Y. Simultaneous Detection of Nine Key Bacterial Respiratory Pathogens Using Luminex xTAG ® Technology. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14030223. [PMID: 28241513 PMCID: PMC5369059 DOI: 10.3390/ijerph14030223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/13/2017] [Accepted: 02/20/2017] [Indexed: 01/26/2023]
Abstract
Early diagnosis and treatment are crucial to the outcome of lower respiratory tract infections (LRTIs). In this study, we developed an assay combining multiplex PCR and Luminex technology (MPLT) for the detection of nine important respiratory bacterial pathogens, which frequently cause LRTIs. These were Streptococcus pneumoniae, Moraxella catarrhalis, Staphylococcus aureus, Streptococcus pyogenes, Haemophilus influenzae, Mycoplasma pneumoniae, Legionella spp., Pseudomonas aeruginosa, and Klebsiella pneumoniae. Through the hybridization reaction between two new synthesized multiplex PCR products and MagPlex-TAG Microspheres, we demonstrate that the detection limits for these nine pathogens were as low as 102–103 CFU/mL. Furthermore, 86 clinical bronchoalveolar lavage fluid specimens were used to evaluate this method. Compared with the results of nine simplex real-time PCR reactions targeting these nine pathogens, this MPLT assay demonstrated a high diagnostic accuracy for Streptococcus pneumoniae (sensitivity, 87.5% and specificity, 100%). Furthermore, sensitivity and specificity for the other eight pathogens all attained 100% diagnostic accuracy. In addition, the consistency between MPLT and the nine real-time PCR reactions exceeded 98.8%. In conclusion, MPLT is a high-throughput, labor-saving and reliable method with high sensitivity and specificity for identifying nine respiratory pathogens responsible for LRTIs. Indeed, this assay may be a promising supplement to conventional methods used to diagnose LRTIs.
Collapse
Affiliation(s)
- Luxi Jiang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
- Department of Respiratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou 310014, China.
| | - Hongyu Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Haijian Zhou
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Tian Qin
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Yu Chen
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
35
|
Stentzel S, Hagl B, Abel F, Kahl BC, Rack-Hoch A, Bröker BM, Renner ED. Reduced Immunoglobulin (Ig) G Response to Staphylococcus aureus in STAT3 Hyper-IgE Syndrome. Clin Infect Dis 2017; 64:1279-1282. [DOI: 10.1093/cid/cix140] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/25/2017] [Indexed: 11/13/2022] Open
|
36
|
Schmidt F, Meyer T, Sundaramoorthy N, Michalik S, Surmann K, Depke M, Dhople V, Gesell Salazar M, Holtappels G, Zhang N, Bröker BM, Bachert C, Völker U. Characterization of human and Staphylococcus aureus proteins in respiratory mucosa by in vivo- and immunoproteomics. J Proteomics 2017; 155:31-39. [DOI: 10.1016/j.jprot.2017.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/28/2016] [Accepted: 01/13/2017] [Indexed: 10/20/2022]
|
37
|
Innate Immune Memory Contributes to Host Defense against Recurrent Skin and Skin Structure Infections Caused by Methicillin-Resistant Staphylococcus aureus. Infect Immun 2017; 85:IAI.00876-16. [PMID: 27849182 DOI: 10.1128/iai.00876-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
Staphylococcus aureus is the leading cause of skin and skin structure infections (SSSI). The high frequency of recurring SSSI due to S. aureus, including methicillin-resistant S. aureus (MRSA) strains, despite high titers of specific antibodies and circulating T cells, implies that traditional adaptive immunity imparts incomplete protection. We hypothesized that innate immune memory contributes to the protective host defense against recurring MRSA infection. To test this hypothesis, SSSI was induced in wild-type and rag1-/- mice in the BALB/c and C57BL/6 backgrounds. Prior infection (priming) of wild-type and rag1-/- mice of either background afforded protection against repeat infection, as evidenced by reduced abscess severities and decreased CFU densities compared to those in naive controls. Interestingly, protection was greater on the previously infected flank than on the naive flank for wild-type and rag1-/- mice. For wild-type mice, protective efficacy corresponded to increased infiltration of neutrophils (polymorphonuclear leukocytes [PMN]), macrophages (MΦ), Langerin+ dendritic cells (LDC), and natural killer (NK) cells. Protection was associated with the induction of interleukin-17A (IL-17A), IL-22, and gamma interferon (IFN-γ) as well as the antimicrobial peptides CRAMP and mβD-3. Priming also protected rag1-/- mice against recurring SSSI, with increased MΦ and LDC infiltration and induction of IL-22, CRAMP, and mβD-3. These findings suggest that innate immune memory, mediated by specific cellular and molecular programs, likely contributes to the localized host defense in recurrent MRSA SSSI. These insights support the development of targeted immunotherapeutic strategies to address the challenge of MRSA infection.
Collapse
|
38
|
Partial Immunoblotting of 2D-Gels: A Novel Method to Identify Post-Translationally Modified Proteins Exemplified for the Myelin Acetylome. Proteomes 2017; 5:proteomes5010003. [PMID: 28248254 PMCID: PMC5372224 DOI: 10.3390/proteomes5010003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 11/17/2022] Open
Abstract
Post-translational modifications (PTMs) play a key role in regulating protein function, yet their identification is technically demanding. Here, we present a straightforward workflow to systematically identify post-translationally modified proteins based on two-dimensional gel electrophoresis. Upon colloidal Coomassie staining the proteins are partially transferred, and the investigated PTMs are immunodetected. This strategy allows tracking back the immunopositive antigens to the corresponding spots on the original gel, from which they are excised and mass spectrometrically identified. Candidate proteins are validated on the same membrane by immunodetection using a second fluorescence channel. We exemplify the power of partial immunoblotting with the identification of lysine-acetylated proteins in myelin, the oligodendroglial membrane that insulates neuronal axons. The excellent consistency of the detected fluorescence signals at all levels allows the differential comparison of PTMs across multiple conditions. Beyond PTM screening, our multi-level workflow can be readily adapted to clinical applications such as identifying auto-immune antigens or host-pathogen interactions.
Collapse
|
39
|
Jorge AM, Schneider J, Unsleber S, Göhring N, Mayer C, Peschel A. Utilization of glycerophosphodiesters by Staphylococcus aureus. Mol Microbiol 2016; 103:229-241. [PMID: 27726204 DOI: 10.1111/mmi.13552] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2016] [Indexed: 01/02/2023]
Abstract
The facultative pathogen Staphylococcus aureus colonizes the human anterior nares and causes infections of various organ systems. Which carbon, energy, and phosphate sources can be utilized by S. aureus in nutrient-poor habitats has remained largely unknown. We describe that S. aureus secretes a glycerophosphodiesterase (glycerophosphodiester phosphodiesterase, EC 3.1.4.46), GlpQ, degrading the glycerophosphodiester (GPD) head groups of phospholipids such as human phosphatidylcholine (GroPC). Deletion of glpQ completely abolished the GroPC-degrading activity in S. aureus culture supernatants. GroPC has been detected in human tissues and body fluids probably as a result of phospholipid remodelling and degradation. Notably, GroPC promoted S. aureus growth under carbon- and phosphate-limiting conditions in a GlpQ-dependent manner indicating that GlpQ permits S. aureus to utilize GPD-derived glycerol-3-phosphate as a carbon and phosphate sources. Thus, S. aureus can use a broader spectrum of nutrients than previously thought which underscores its capacity to adapt to the highly variable and nutrient-poor surroundings.
Collapse
Affiliation(s)
- Ana Maria Jorge
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Jonathan Schneider
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Sandra Unsleber
- Interfaculty Institute of Microbiology and Infection Medicine, Microbiology/Biotechnology Department, University of Tübingen, Germany
| | - Nadine Göhring
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Christoph Mayer
- Interfaculty Institute of Microbiology and Infection Medicine, Microbiology/Biotechnology Department, University of Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| |
Collapse
|
40
|
Stentzel S, Gläser R, Bröker BM. Elucidating the anti-Staphylococcus aureusantibody response by immunoproteomics. Proteomics Clin Appl 2016; 10:1011-1019. [DOI: 10.1002/prca.201600050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/20/2016] [Accepted: 08/08/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| | - Regine Gläser
- Department of Dermatology; University Hospital Schleswig-Holstein; Kiel Germany
| | - Barbara M. Bröker
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| |
Collapse
|
41
|
Combining in vitro protein detection and in vivo antibody detection identifies potential vaccine targets against Staphylococcus aureus during osteomyelitis. Med Microbiol Immunol 2016; 206:11-22. [PMID: 27629411 PMCID: PMC5263195 DOI: 10.1007/s00430-016-0476-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/03/2016] [Indexed: 12/31/2022]
Abstract
Currently, little is known about the in vivo human immune response against Staphylococcus aureus during a biofilm-associated infection, such as osteomyelitis, and how this relates to protein production in biofilms in vitro. Therefore, we characterized IgG responses in 10 patients with chronic osteomyelitis against 50 proteins of S. aureus, analyzed the presence of these proteins in biofilms of the infecting isolates on polystyrene (PS) and human bone in vitro, and explored the relation between in vivo and in vitro data. IgG levels against 15 different proteins were significantly increased in patients compared to healthy controls. Using a novel competitive Luminex-based assay, eight of these proteins [alpha toxin, Staphylococcus aureus formyl peptide receptor-like 1 inhibitor (FlipR), glucosaminidase, iron-responsive surface determinants A and H, the putative ABC transporter SACOL0688, staphylococcal complement inhibitor (SCIN), and serine-aspartate repeat-containing protein E (SdrE)] were also detected in a majority of the infecting isolates during biofilm formation in vitro. However, 4 other proteins were detected in only a minority of isolates in vitro while, vice versa, 7 proteins were detected in multiple isolates in vitro but not associated with significantly increased IgG levels in patients. Detection of proteins was largely confirmed using a transcriptomic approach. Our data provide further insights into potential therapeutic targets, such as for vaccination, to reduce S. aureus virulence and biofilm formation. At the same time, our data suggest that either in vitro or immunological in vivo data alone should be interpreted cautiously and that combined studies are necessary to identify potential targets.
Collapse
|
42
|
Stentzel S, Teufelberger A, Nordengrün M, Kolata J, Schmidt F, van Crombruggen K, Michalik S, Kumpfmüller J, Tischer S, Schweder T, Hecker M, Engelmann S, Völker U, Krysko O, Bachert C, Bröker BM. Staphylococcal serine protease-like proteins are pacemakers of allergic airway reactions to Staphylococcus aureus. J Allergy Clin Immunol 2016; 139:492-500.e8. [PMID: 27315768 DOI: 10.1016/j.jaci.2016.03.045] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/15/2016] [Accepted: 03/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND A substantial subgroup of asthmatic patients have "nonallergic" or idiopathic asthma, which often takes a severe course and is difficult to treat. The cause might be allergic reactions to the gram-positive pathogen Staphylococcus aureus, a frequent colonizer of the upper airways. However, the driving allergens of S aureus have remained elusive. OBJECTIVE We sought to search for potentially allergenic S aureus proteins and characterize the immune response directed against them. METHODS S aureus extracellular proteins targeted by human serum IgG4 were identified by means of immunoblotting to screen for potential bacterial allergens. Candidate antigens were expressed as recombinant proteins and used to analyze the established cellular and humoral immune responses in healthy adults and asthmatic patients. The ability to induce a type 2 immune response in vivo was tested in a mouse asthma model. RESULTS We identified staphylococcal serine protease-like proteins (Spls) as dominant IgG4-binding S aureus proteins. SplA through SplF are extracellular proteases of unknown function expressed by S aureus in vivo. Spls elicited IgE antibody responses in most asthmatic patients. In healthy S aureus carriers and noncarriers, peripheral blood T cells elaborated TH2 cytokines after stimulation with Spls, as is typical for allergens. In contrast, TH1/TH17 cytokines, which dominated the response to S aureus α-hemolysin, were of low concentration or absent. In mice inhalation of SplD without adjuvant induced lung inflammation characterized by TH2 cytokines and eosinophil infiltration. CONCLUSION We identify Spls as triggering allergens released by S aureus, opening prospects for diagnosis and causal therapy of asthma.
Collapse
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Julia Kolata
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany; Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank Schmidt
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | | | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | - Jana Kumpfmüller
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, HKI, Jena, Germany
| | - Sebastian Tischer
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Schweder
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Michael Hecker
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Susanne Engelmann
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Institute for Microbiology, University of Braunschweig, Braunschweig, Germany; Helmholtz Center for Infection Research, Microbial Proteomics, Braunschweig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Olga Krysko
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium; Division of Ear, Nose, and Throat Diseases, Clintec, Karolinska Institute, Stockholm, Sweden
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
43
|
Bermejo-Martin JF, Andaluz-Ojeda D, Almansa R, Gandía F, Gómez-Herreras JI, Gomez-Sanchez E, Heredia-Rodríguez M, Eiros JM, Kelvin DJ, Tamayo E. Defining immunological dysfunction in sepsis: A requisite tool for precision medicine. J Infect 2016; 72:525-36. [PMID: 26850357 DOI: 10.1016/j.jinf.2016.01.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/24/2016] [Accepted: 01/26/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Immunological dysregulation is now recognised as a major pathogenic event in sepsis. Stimulation of immune response and immuno-modulation are emerging approaches for the treatment of this disease. Defining the underlying immunological alterations in sepsis is important for the design of future therapies with immuno-modulatory drugs. METHODS Clinical studies evaluating the immunological response in adult patients with Sepsis and published in PubMed were reviewed to identify features of immunological dysfunction. For this study we used key words related with innate and adaptive immunity. RESULTS Ten major features of immunological dysfunction (FID) were identified involving quantitative and qualitative alterations of [antigen presentation](FID1), [T and B lymphocytes] (FID2), [natural killer cells] (FID3), [relative increase in T regulatory cells] (FID4), [increased expression of PD-1 and PD-ligand1](FID5), [low levels of immunoglobulins](FID6), [low circulating counts of neutrophils and/or increased immature forms in non survivors](FID7), [hyper-cytokinemia] (FID8), [complement consumption] (FID9), [defective bacterial killing by neutrophil extracellular traps](FID10). CONCLUSIONS This review article identified ten major features associated with immunosuppression and immunological dysregulation in sepsis. Assessment of these features could help in utilizing precision medicine for the treatment of sepsis with immuno-modulatory drugs.
Collapse
Affiliation(s)
- Jesús F Bermejo-Martin
- Infection and Immunity Medical Investigation Unit (IMI), Hospital Clínico Universitario de Valladolid, SACYL/IECSCYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - David Andaluz-Ojeda
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Medicina Intensiva, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Raquel Almansa
- Infection and Immunity Medical Investigation Unit (IMI), Hospital Clínico Universitario de Valladolid, SACYL/IECSCYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Francisco Gandía
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Medicina Intensiva, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jose Ignacio Gómez-Herreras
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Esther Gomez-Sanchez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - María Heredia-Rodríguez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jose Maria Eiros
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - David J Kelvin
- Division of Experimental Therapeutics, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada; Sezione di Microbiologia Sperimentale e Clinica, Dipartimento di Scienze Biomediche, Universita' degli Studi di Sassari, Piazza Università, 21, 07100 Sassari SS, Italy; International Institute of Infection and Immunity, Shantou University Medical College, 22 Xinling Road, Shantou, 515041 Guangdong Province, PR China.
| | - Eduardo Tamayo
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| |
Collapse
|
44
|
Selle M, Hertlein T, Oesterreich B, Klemm T, Kloppot P, Müller E, Ehricht R, Stentzel S, Bröker BM, Engelmann S, Ohlsen K. Global antibody response to Staphylococcus aureus live-cell vaccination. Sci Rep 2016; 6:24754. [PMID: 27103319 PMCID: PMC4840433 DOI: 10.1038/srep24754] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/05/2016] [Indexed: 02/06/2023] Open
Abstract
The pathogen Staphylococcus aureus causes a broad range of severe diseases and is feared for its ability to rapidly develop resistance to antibiotic substances. The increasing number of highly resistant S. aureus infections has accelerated the search for alternative treatment options to close the widening gap in anti-S. aureus therapy. This study analyses the humoral immune response to vaccination of Balb/c mice with sublethal doses of live S. aureus. The elicited antibody pattern in the sera of intravenously and intramuscularly vaccinated mice was determined using of a recently developed protein array. We observed a specific antibody response against a broad set of S. aureus antigens which was stronger following i.v. than i.m. vaccination. Intravenous but not intramuscular vaccination protected mice against an intramuscular challenge infection with a high bacterial dose. Vaccine protection was correlated with the strength of the anti-S. aureus antibody response. This study identified novel vaccine candidates by using protein microarrays as an effective tool and showed that successful vaccination against S. aureus relies on the optimal route of administration.
Collapse
Affiliation(s)
- Martina Selle
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Tobias Hertlein
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Babett Oesterreich
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Theresa Klemm
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Peggy Kloppot
- University Greifswald, Institute for Microbiology, Greifswald, Germany
| | - Elke Müller
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Ralf Ehricht
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Sebastian Stentzel
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Barbara M Bröker
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Susanne Engelmann
- Technical University Braunschweig, Institute for Microbiology, Braunschweig, Germany.,Helmholtz-Zentrum für Infektionsforschung, Mikrobielle Proteomik, Braunschweig, Germany
| | - Knut Ohlsen
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| |
Collapse
|
45
|
Omics Approaches for the Study of Adaptive Immunity to Staphylococcus aureus and the Selection of Vaccine Candidates. Proteomes 2016; 4:proteomes4010011. [PMID: 28248221 PMCID: PMC5217363 DOI: 10.3390/proteomes4010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/05/2016] [Accepted: 03/01/2016] [Indexed: 01/20/2023] Open
Abstract
Staphylococcus aureus is a dangerous pathogen both in hospitals and in the community. Due to the crisis of antibiotic resistance, there is an urgent need for new strategies to combat S. aureus infections, such as vaccination. Increasing our knowledge about the mechanisms of protection will be key for the successful prevention or treatment of S. aureus invasion. Omics technologies generate a comprehensive picture of the physiological and pathophysiological processes within cells, tissues, organs, organisms and even populations. This review provides an overview of the contribution of genomics, transcriptomics, proteomics, metabolomics and immunoproteomics to the current understanding of S. aureus‑host interaction, with a focus on the adaptive immune response to the microorganism. While antibody responses during colonization and infection have been analyzed in detail using immunoproteomics, the full potential of omics technologies has not been tapped yet in terms of T-cells. Omics technologies promise to speed up vaccine development by enabling reverse vaccinology approaches. In consequence, omics technologies are powerful tools for deepening our understanding of the “superbug” S. aureus and for improving its control.
Collapse
|
46
|
Immunomodulation and Disease Tolerance to Staphylococcus aureus. Pathogens 2015; 4:793-815. [PMID: 26580658 PMCID: PMC4693165 DOI: 10.3390/pathogens4040793] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022] Open
Abstract
The Gram-positive bacterium Staphylococcus aureus is one of the most frequent pathogens that causes severe morbidity and mortality throughout the world. S. aureus can infect skin and soft tissues or become invasive leading to diseases such as pneumonia, endocarditis, sepsis or toxic shock syndrome. In contrast, S. aureus is also a common commensal microbe and is often part of the human nasal microbiome without causing any apparent disease. In this review, we explore the immunomodulation and disease tolerance mechanisms that promote commensalism to S. aureus.
Collapse
|