1
|
Bell RD, Cann EA, Mishra B, Valencia M, Zhang Q, Huang M, Yang X, Carli A, Bostrom M, Ivashkiv LB. Staphyloccocus aureus biofilm, in absence of planktonic bacteria, produces factors that activate counterbalancing inflammatory and immune-suppressive genes in human monocytes. J Orthop Res 2024; 42:2582-2592. [PMID: 38922976 PMCID: PMC11481048 DOI: 10.1002/jor.25919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/25/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Staphyloccocus aureus (S. aureus) is a major bacterial pathogen in orthopedic periprosthetic joint infection (PJI). S. aureus forms biofilms that promote persistent infection by shielding bacteria from immune cells and inducing an antibiotic-tolerant metabolic state. We developed an in vitro system to study S. aureus biofilm interactions with primary human monocytes in the absence of planktonic bacteria. In line with previous in vivo data, S. aureus biofilm induced expression of inflammatory genes such as TNF and IL1B, and their anti-inflammatory counter-regulator IL10. S. aureus biofilm also activated expression of PD-1 ligands, and IL-1RA, molecules that have the potential to suppress T cell function or differentiation of protective Th17 cells. Gene induction did not require monocyte:biofilm contact and was mediated by a soluble factor(s) produced by biofilm-encased bacteria that was heat resistant and >3 kD in size. Activation of suppressive genes by biofilm was sensitive to suppression by Jak kinase inhibition. These results support an evolving paradigm that biofilm plays an active role in modulating immune responses, and suggest this occurs via production of a soluble vita-pathogen-associated molecular pattern, a molecule that signals microbial viability. Induction of T cell suppressive genes by S. aureus biofilm provides insights into mechanisms that can suppress T cell immunity in PJI.
Collapse
Affiliation(s)
- Richard D Bell
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - E. Abrefi Cann
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Bikash Mishra
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine
| | - Melanie Valencia
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Qiong Zhang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Mary Huang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Xu Yang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Alberto Carli
- Department of Orthopedic Surgery, Hospital for Special Surgery
| | - Mathias Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery
| | - Lionel B Ivashkiv
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine
| |
Collapse
|
2
|
Walter N, Bärtl S, Alt V, Rupp M. Recent advancements and future directions in fracture related infections: A scoping review. Injury 2024; 55 Suppl 6:111902. [PMID: 39482033 DOI: 10.1016/j.injury.2024.111902] [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: 03/21/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 11/03/2024]
Abstract
Fracture-related infections (FRIs) are complex challenges in orthopedic and trauma surgery, driving ongoing advancements in diagnostics, therapeutics, and management strategies. This scoping review examines recent progress and future directions in FRI management. Diagnostic enhancements encompass standardized definitions, improved biomarkers, advanced microbiological techniques, and innovative imaging modalities. Promising future diagnostics may include point-of-care testing, advanced imaging with enhanced specificity, and machine learning algorithms. Advancements in implant technology emphasize materials science, surface modifications, and personalized 3D printing, enhancing durability and antimicrobial efficacy. Immunomodulatory therapies targeting T cell dysfunction offer potential in addressing FRI chronicity. Enzybiotics and phages present promising alternatives to combat antibiotic resistance, with enzybiotics demonstrating effectiveness against biofilm-associated infections. Patient optimization, multidisciplinary approaches and specialized reference centers play vital roles in comprehensive FRI management, particularly crucial in resource-constrained settings. Collaboration and investment in research and technology are imperative for harnessing the full potential of these advancements and improving global FRI management outcomes. Addressing these complexities necessitates a multifaceted approach integrating clinical expertise, technological innovation, and global cooperation to optimize patient care and mitigate the burden of FRI worldwide.
Collapse
Affiliation(s)
- Nike Walter
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany.
| | - Susanne Bärtl
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Volker Alt
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Markus Rupp
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| |
Collapse
|
3
|
Van Roy Z, Arumugam P, Bertrand BP, Shinde DD, Thomas VC, Kielian T. Tissue niche influences immune and metabolic profiles to Staphylococcus aureus biofilm infection. Nat Commun 2024; 15:8965. [PMID: 39420209 PMCID: PMC11487069 DOI: 10.1038/s41467-024-53353-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Infection is a devastating post-surgical complication, often requiring additional procedures and prolonged antibiotic therapy. This is especially relevant for craniotomy and prosthetic joint infections (PJI), both of which are characterized by biofilm formation on the bone or implant surface, respectively, with S. aureus representing a primary cause. The local tissue microenvironment likely has profound effects on immune attributes that can influence treatment efficacy, which becomes critical to consider when developing therapeutics for biofilm infections. However, the extent to which distinct tissue niches influence immune function during biofilm development remains relatively unknown. To address this, we compare the metabolomic, transcriptomic, and functional attributes of leukocytes in mouse models of S. aureus craniotomy and PJI complemented with patient samples from both infection modalities, which reveals profound tissue niche-dependent differences in nucleic acid, amino acid, and lipid metabolism with links to immune modulation. These signatures are both spatially and temporally distinct, differing not only between infection sites but evolving over time within a single model. Collectively, this demonstrates that biofilms elicit unique immune and metabolic responses that are heavily influenced by the local tissue microenvironment, which will likely have important implications when designing therapeutic approaches targeting these infections.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prabakar Arumugam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Blake P Bertrand
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dhananjay D Shinde
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vinai C Thomas
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Argyrou C, Papagrigorakis E, Tzefronis D, Pliaka V, Fotis C, Kamariotis S, Chatzinikolaidou M, Tsiamtsouris K, Vasiliadis ES, Alexopoulos L, Macheras GA. Multiplex cytokine analysis for the identification of novel potential synovial fluid biomarkers for periprosthetic joint infections. Injury 2024; 55:111659. [PMID: 38917741 DOI: 10.1016/j.injury.2024.111659] [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: 03/02/2024] [Revised: 05/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Periprosthetic joint infections (PJIs) are a devastating complication of total hip (THA) and knee (TKA) arthroplasty. The use of novel techniques like multiplex cytokine analysis could contribute immensely to the identification of potential novel biomarkers. PATIENTS AND METHODS This is a single-centre study of patients that were treated with revision TKA, THA or hemiarthroplasty. Serum's white blood cells (WBCs), erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) and synovial fluid's WBCs, percentage of polymorphonuclear neutrophils (%PMNs) and CRP were measured. Proteomic analysis targeting the secreted cytokines in synovial fluid was conducted using a 73-plex assay panel. The results were statistically compared between the septic and aseptic cases and ROC analysis to establish the area under the curve (AUC), sensitivity and specificity of each biomarker. RESULTS The study included 30 patients (18 revision THA cases; 3 conversion of hemiarthroplasty to THA and 9 revision TKA cases); 14 cases were considered infected, 1 likely infected and 15 not infected. The results showed statistically significant differences (p < 0.05) between infected and not infected cases in serum's ESR, CRP and synovial fluid's%PMNs, growth-regulated oncogene alpha (GROA), interleukin-8, interleukin-5, S100-A8/calprotectin and resistin (RETN) with AUCs of 0.75, 0.72, 0.95, 0.75, 0.72, 0.95, 0.83, 0.73, 0.75, 0.81 and 0.76 respectively. CONCLUSIONS In the present study, serum ESR and CRP as well as synovial %PMNs, GROA, IL-8, IL-5, calprotectin and RETN protein levels were identified as potential biomarkers. Further studies are needed to further investigate their diagnostic utility and optimal cut-off values.
Collapse
Affiliation(s)
- Chrysoula Argyrou
- 4th Department of Orthopaedics, KAT Attica General Hospital, Athens, Greece.
| | - Eftychios Papagrigorakis
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Attica General Hospital, Athens, Greece
| | - Dimitrios Tzefronis
- 7th Department of Orthopaedic Surgery, Henry Dunant Hospital Center, Athens, Greece
| | - Vaia Pliaka
- Biotechnology, Protavio Ltd., Athens, Greece
| | | | - Spyros Kamariotis
- Microbiology Department, KAT Attica General Hospital, Athens, Greece
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece & FORTH-IESL, 70013 Heraklion, Greece
| | | | - Elias S Vasiliadis
- 3rd Department of Orthopaedics, School of Medicine, National and Kapodistrian University of Athens, KAT Attica General Hospital, Athens, Greece
| | - Leonidas Alexopoulos
- Biotechnology, Protavio Ltd., Athens, Greece; School of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | - George A Macheras
- 4th Department of Orthopaedics, KAT Attica General Hospital, Athens, Greece; 7th Department of Orthopaedic Surgery, Henry Dunant Hospital Center, Athens, Greece
| |
Collapse
|
5
|
Korshoj LE, Kielian T. Bacterial single-cell RNA sequencing captures biofilm transcriptional heterogeneity and differential responses to immune pressure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601229. [PMID: 38979200 PMCID: PMC11230364 DOI: 10.1101/2024.06.28.601229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Biofilm formation is an important mechanism of survival and persistence for many bacterial pathogens. These multicellular communities contain subpopulations of cells that display vast metabolic and transcriptional diversity along with high recalcitrance to antibiotics and host immune defenses. Investigating the complex heterogeneity within biofilm has been hindered by the lack of a sensitive and high-throughput method to assess stochastic transcriptional activity and regulation between bacterial subpopulations, which requires single-cell resolution. We have developed an optimized bacterial single-cell RNA sequencing method, BaSSSh-seq, to study Staphylococcus aureus diversity during biofilm growth and transcriptional adaptations following immune cell exposure. We validated the ability of BaSSSh-seq to capture extensive transcriptional heterogeneity during biofilm compared to planktonic growth. Application of new computational tools revealed transcriptional regulatory networks across the heterogeneous biofilm subpopulations and identification of gene sets that were associated with a trajectory from planktonic to biofilm growth. BaSSSh-seq also detected alterations in biofilm metabolism, stress response, and virulence that were tailored to distinct immune cell populations. This work provides an innovative platform to explore biofilm dynamics at single-cell resolution, unlocking the potential for identifying biofilm adaptations to environmental signals and immune pressure.
Collapse
|
6
|
Deans CF, Kildow BJ, Garvin KL. Recurrent Periprosthetic Joint Infections: Diagnosis, Management, and Outcomes. Orthop Clin North Am 2024; 55:193-206. [PMID: 38403366 DOI: 10.1016/j.ocl.2023.09.002] [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] [Indexed: 02/27/2024]
Abstract
Periprosthetic joint infection (PJI) remains one of the most common complications after total joint arthroplasty. It is challenging to manage, associated with significant morbidity and mortality, and is a financial burden on the health care system. Failure of 2-stage management for chronic PJI is not uncommon. Repeat infections are oftentimes polymicrobial, multiple drug-resistant microorganisms, or new organisms. Optimizing the success of index 2-stage revision is the greatest prevention against failure of any subsequent management options and requires a robust team-based approach.
Collapse
Affiliation(s)
- Christopher F Deans
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, 985640 Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Beau J Kildow
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, 985640 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kevin L Garvin
- Department of Orthopaedic Surgery and Rehabilitation, University of Nebraska Medical Center, 985640 Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
7
|
Mitterer JA, Frank BJH, Sebastian S, Guger M, Schoefberger L, Hofstaetter JG. The Value of Preoperative Ultrasound-Determined Fluid Film and Joint Aspiration in Revision Hip Arthroplasty. J Arthroplasty 2024; 39:1069-1074. [PMID: 37866461 DOI: 10.1016/j.arth.2023.10.029] [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: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Data regarding the diagnostic value of ultrasound (US)-determined fluid film and joint aspiration prior to revision total hip arthroplasty for suspected periprosthetic joint infections (PJIs) are limited. This study aimed to analyze the value of US-determined fluid film, characterized the preoperative and intraoperative microbiological spectrum and resistance patterns, and compared the concordance between preoperative synovial fluid and intraoperative culture results. METHODS We analyzed 366 US examinations from 324 patients prior to revision total hip arthroplasty. Selected cases were grouped into clearly infected, noninfected, and inconclusive cohorts, according to the International Consensus Meeting 2018 Criteria. For US-determined fluid film <1 mm, no aspiration was performed based on our institutional protocol. Patients were grouped into no aspiration (144 of 366; [39.3%]), dry tap (21 of 366; [5.7%]), and a successful tap (201 of 366; [54.9%]). The microbiological spectrum and antibiograms were compared between preoperative and intraoperative results. RESULTS The absence of US-determined fluid film showed no correlation with the presence of a hip PJI. Overall, 31.9% cases of the no-aspiration group had a PJI. In total, 13.5% discrepancies were found between successful taps and intraoperative cultures. The most prevalent microorganisms in preoperative synovial fluid were Staphylococcus epidermidis and Staphylococcus aureus (20.8%), while intraoperatively S. epidermidis (26.3%) and Cutibacterium acnes (14.5%) were leading. Additional microorganisms were identified in 32.5% of intraoperative cultures. There were no differences between resistance patterns of preoperative and intraoperative concordant microorganisms. CONCLUSIONS Absence of US-determined fluid film cannot rule out the presence of a hip PJI. Combined microbiological results from hip US aspirations and subsequent surgical procedures are crucial to design an effective treatment for suspected hip PJI.
Collapse
Affiliation(s)
- Jennyfer A Mitterer
- Michael Ogon Laboratory for Orthopaedic Research, Orthopaedic Hospital Speising, Vienna, Austria
| | - Bernhard J H Frank
- Michael Ogon Laboratory for Orthopaedic Research, Orthopaedic Hospital Speising, Vienna, Austria; 2nd Department, Orthopaedic Hospital Speising, Vienna, Austria
| | - Sujeesh Sebastian
- Michael Ogon Laboratory for Orthopaedic Research, Orthopaedic Hospital Speising, Vienna, Austria
| | - Matthias Guger
- Institute for Radiology, Orthopaedic Hospital Speising, Vienna, Austria
| | - Lukas Schoefberger
- Michael Ogon Laboratory for Orthopaedic Research, Orthopaedic Hospital Speising, Vienna, Austria
| | - Jochen G Hofstaetter
- Michael Ogon Laboratory for Orthopaedic Research, Orthopaedic Hospital Speising, Vienna, Austria; 2nd Department, Orthopaedic Hospital Speising, Vienna, Austria
| |
Collapse
|
8
|
Akaraphanth M, Nordgren TM, Gries CM. CXCR2 perturbation promotes Staphylococcus aureus implant-associated infection. J Med Microbiol 2024; 73:001821. [PMID: 38567642 PMCID: PMC11084549 DOI: 10.1099/jmm.0.001821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction. Staphylococcus aureus is the leading cause of acute medical implant infections, representing a significant modern medical concern. The success of S. aureus as a pathogen in these cases resides in its arsenal of virulence factors, resistance to multiple antimicrobials, mechanisms of immune modulation, and ability to rapidly form biofilms associated with implant surfaces. S. aureus device-associated, biofilm-mediated infections are often persistent and notoriously difficult to treat, skewing innate immune responses to promote chronic reoccurring infections. While relatively little is known of the role neutrophils play in response to acute S. aureus biofilm infections, these effector cells must be efficiently recruited to sites of infection via directed chemotaxis. Here we investigate the effects of modulating CXC chemokine receptor 2 (CXCR2) activity, predominantly expressed on neutrophils, during S. aureus implant-associated infection.Hypothesis. We hypothesize that modulation of CXCR2 expression and/or signalling activities during S. aureus infection, and thus neutrophil recruitment, extravasation and antimicrobial activity, will affect infection control and bacterial burdens in a mouse model of implant-associated infection.Aim. This investigation aims to elucidate the impact of altered CXCR2 activity during S. aureus biofilm-mediated infection that may help develop a framework for an effective novel strategy to prevent morbidity and mortality associated with implant infections.Methodology. To examine the role of CXCR2 during S. aureus implant infection, we employed a mouse model of indwelling subcutaneous catheter infection using a community-associated methicillin-resistant S. aureus (MRSA) strain. To assess the role of CXCR2 induction or inhibition during infection, treatment groups received daily intraperitoneal doses of either Lipocalin-2 (Lcn2) or AZD5069, respectively. At the end of the study, catheters and surrounding soft tissues were analysed for bacterial burdens and dissemination, and Cxcr2 transcription within the implant-associated tissues was quantified.Results. Mice treated with Lcn2 developed higher bacterial burdens within the soft tissue surrounding the implant site, which was associated with increased Cxcr2 expression. AZD5069 treatment also resulted in increased implant- and tissues-associated bacterial titres, as well as enhanced Cxcr2 expression.Conclusion. Our results demonstrate that CXCR2 plays an essential role in regulating the severity of S. aureus implant-associated infections. Interestingly, however, perturbation of CXCR2 expression or signalling both resulted in enhanced Cxcr2 transcription and elevated implant-associated bacterial burdens. Thus, CXCR2 appears finely tuned to efficiently recruit effector cells and mediate control of S. aureus biofilm-mediated infection.
Collapse
Affiliation(s)
- Mike Akaraphanth
- School of Medicine, University of Colorado, Aurora CO 80045, USA
| | - Tara M. Nordgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins CO 80523, USA
| | - Casey M. Gries
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins CO 80523, USA
| |
Collapse
|
9
|
Bertrand BP, Heim CE, Koepsell SA, Kielian T. Elucidating granulocytic myeloid-derived suppressor cell heterogeneity during Staphylococcus aureus biofilm infection. J Leukoc Biol 2024; 115:620-632. [PMID: 38095415 DOI: 10.1093/jleuko/qiad158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 03/02/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are pathologically activated immature myeloid cells with immunosuppressive activity that expand during chronic inflammation, such as cancer and prosthetic joint infection (PJI). Myeloid-derived suppressor cells can be broadly separated into 2 populations based on surface marker expression and function: monocytic myeloid-derived suppressor cells (M-MDSCs) and granulocytic myeloid-derived suppressor cells (G-MDSCs). Granulocytic myeloid-derived suppressor cells are the most abundant leukocyte infiltrate during PJI; however, how this population is maintained in vivo and cellular heterogeneity is currently unknown. In this study, we identified a previously unknown population of Ly6G+Ly6C+F4/80+MHCII+ MDSCs during PJI that displayed immunosuppressive properties ex vivo. We leveraged F4/80 and MHCII expression by these cells for further characterization using cellular indexing of transcriptomes and epitopes by sequencing, which revealed a distinct transcriptomic signature of this population. F4/80+MHCII+ MDSCs displayed gene signatures resembling G-MDSCs, neutrophils, and monocytes but had significantly increased expression of pathways involved in cytokine response/production, inflammatory cell death, and mononuclear cell differentiation. To determine whether F4/80+MHCII+ MDSCs represented an alternate phenotypic state of G-MDSCs, Ly6G+Ly6C+F4/80-MHCII- G-MDSCs from CD45.1 mice were adoptively transferred into CD45.2 recipients using a mouse model of PJI. A small percentage of transferred G-MDSCs acquired F4/80 and MHCII expression in vivo, suggesting some degree of plasticity in this population. Collectively, these results demonstrate a previously unappreciated phenotype of F4/80+MHCII+ MDSCs during PJI, revealing that a granulocytic-to-monocytic transition can occur during biofilm infection.
Collapse
Affiliation(s)
- Blake P Bertrand
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, United States
| | - Cortney E Heim
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, United States
| | - Scott A Koepsell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, United States
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, United States
| |
Collapse
|
10
|
Jiang F, Wang J, Ren Z, Hu Y, Wang B, Li M, Yu J, Tang J, Guo G, Cheng Y, Han P, Shen H. Targeted Light-Induced Immunomodulatory Strategy for Implant-Associated Infections via Reversing Biofilm-Mediated Immunosuppression. ACS NANO 2024; 18:6990-7010. [PMID: 38385433 DOI: 10.1021/acsnano.3c10172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The clinical treatment efficacy for implant-associated infections (IAIs), particularly those caused by Methicillin-resistant Staphylococcus aureus (MRSA), remains unsatisfactory, primarily due to the formation of biofilm barriers and the resulting immunosuppressive microenvironment, leading to the chronicity and recurrence of IAIs. To address this challenge, we propose a light-induced immune enhancement strategy, synthesizing BSA@MnO2@Ce6@Van (BMCV). The BMCV exhibits precise targeting and adhesion to the S. aureus biofilm-infected region, coupled with its capacity to catalyze oxygen generation from H2O2 in the hypoxic and acidic biofilm microenvironment (BME), promoting oxygen-dependent photodynamic therapy efficacy while ensuring continuous release of manganese ions. Notably, targeted BMCV can penetrate biofilms, producing ROS that degrade extracellular DNA, disrupting the biofilm structure and impairing its barrier function, making it vulnerable to infiltration and elimination by the immune system. Furthermore, light-induced reactive oxygen species (ROS) around the biofilm can lyse S. aureus, triggering bacterium-like immunogenic cell death (ICD), releasing abundant immune costimulatory factors, facilitating the recognition and maturation of antigen-presenting cells (APCs), and activating adaptive immunity. Additionally, manganese ions in the BME act as immunoadjuvants, further amplifying macrophage-mediated innate and adaptive immune responses and reversing the immunologically cold BME to an immunologically hot BME. We prove that our synthesized BMCV elicits a robust adaptive immune response in vivo, effectively clearing primary IAIs and inducing long-term immune memory to prevent recurrence. Our study introduces a potent light-induced immunomodulatory nanoplatform capable of reversing the biofilm-induced immunosuppressive microenvironment and disrupting biofilm-mediated protective barriers, offering a promising immunotherapeutic strategy for addressing challenging S. aureus IAIs.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jian Wang
- Department of Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zun Ren
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yujie Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Boyong Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mingzhang Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Geyong Guo
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yingsheng Cheng
- Department of Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Imaging Medicine and Nuclear Medicine, Tongji Hospital, Shanghai 200065, China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
11
|
Horn CM, Arumugam P, Van Roy Z, Heim CE, Fallet RW, Bertrand BP, Shinde D, Thomas VC, Romanova SG, Bronich TK, Hartman CW, Garvin KL, Kielian T. Granulocytic myeloid-derived suppressor cell activity during biofilm infection is regulated by a glycolysis/HIF1a axis. J Clin Invest 2024; 134:e174051. [PMID: 38421730 PMCID: PMC11014666 DOI: 10.1172/jci174051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 02/20/2024] [Indexed: 03/02/2024] Open
Abstract
Staphylococcus aureus is a leading cause of biofilm-associated prosthetic joint infection (PJI). A primary contributor to infection chronicity is an expansion of granulocytic myeloid-derived suppressor cells (G-MDSCs), which are critical for orchestrating the antiinflammatory biofilm milieu. Single-cell sequencing and bioinformatic metabolic algorithms were used to explore the link between G-MDSC metabolism and S. aureus PJI outcome. Glycolysis and the hypoxia response through HIF1a were significantly enriched in G-MDSCs. Interfering with both pathways in vivo, using a 2-deoxyglucose nanopreparation and granulocyte-targeted Hif1a conditional KO mice, respectively, attenuated G-MDSC-mediated immunosuppression and reduced bacterial burden in a mouse model of S. aureus PJI. In addition, single-cell RNA-Seq (scRNA-Seq) analysis of granulocytes from PJI patients also showed an enrichment in glycolysis and hypoxia-response genes. These findings support the importance of a glycolysis/HIF1a axis in promoting G-MDSC antiinflammatory activity and biofilm persistence during PJI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Svetlana G. Romanova
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Tatiana K. Bronich
- Department of Pharmacy, Northeastern University, Boston, Massachusetts, USA
| | - Curtis W. Hartman
- Department of Orthopaedic Surgery and Rehabilitation, UNMC, Omaha, Nebraska, USA
| | - Kevin L. Garvin
- Department of Orthopaedic Surgery and Rehabilitation, UNMC, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology and
| |
Collapse
|
12
|
Gómez-Alonso IS, Betanzos-Cabrera G, Moreno-Lafont MC, Cancino-Diaz ME, García-Pérez BE, Cancino-Diaz JC. Non-biofilm-forming Staphylococcus epidermidis planktonic cell supernatant induces alterations in osteoblast biological function. Sci Rep 2024; 14:1807. [PMID: 38245549 PMCID: PMC10799936 DOI: 10.1038/s41598-024-51899-7] [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: 10/30/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024] Open
Abstract
Staphylococcal biofilms significantly contribute to prosthetic joint infection (PJI). However, 40% of S. epidermidis PJI isolates do not produce biofilms, which does not explain the role of biofilms in these cases. We studied whether the supernatant from planktonic S. epidermidis alters osteoblast function. Non-biofilm-forming S. epidermidis supernatants (PJI- clinical isolate, healthy skin isolate (HS), and ATCC12228 reference strain) and biofilm-forming supernatants (PJI+ clinical isolate, ATCC35984 reference strain, and Staphylococcus aureus USA300 reference strain) were included. Osteoblasts stimulated with supernatants from non-biofilm-forming isolates for 3, 7, and 14 days showed significantly reduced cellular DNA content compared with unstimulated osteoblasts, and apoptosis was induced in these osteoblasts. Similar results were obtained for biofilm-forming isolates, but with a greater reduction in DNA content and higher apoptosis. Alkaline phosphatase activity and mineralization were significantly reduced in osteoblasts treated with supernatants from non-biofilm-forming isolates compared to the control at the same time points. However, the supernatants from biofilm-forming isolates had a greater effect than those from non-biofilm-forming isolates. A significant decrease in the expression of ATF4, RUNX2, ALP, SPARC, and BGLAP, and a significant increase in RANK-L expression were observed in osteoblasts treated with both supernatants. These results demonstrate that the supernatants of the S. epidermidis isolate from the PJI- and HS (commensal) with a non-biofilm-forming phenotype alter the function of osteoblasts (apoptosis induction, failure of cell differentiation, activation of osteoblasts, and induction of bone resorption), similar to biofilm-forming isolates (PJI+, ATCC35984, and S. aureus USA300), suggesting that biofilm status contributes to impaired osteoblast function and that the planktonic state can do so independently of biofilm production.
Collapse
Affiliation(s)
- Itzia Sidney Gómez-Alonso
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Gabriel Betanzos-Cabrera
- Área Académica de Nutrición, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Actopan Camino a Tilcuautla S/N., Pueblo San Juan Tilcuautla, 42160, Pachuca Hidalgo, Mexico
| | - Martha Cecilia Moreno-Lafont
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Mario Eugenio Cancino-Diaz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Blanca Estela García-Pérez
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico
| | - Juan Carlos Cancino-Diaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Manuel Carpio, Plutarco Elías Calles, Miguel Hidalgo, 11350, Mexico City, Mexico.
| |
Collapse
|
13
|
Piuzzi NS, Klika AK, Lu Q, Higuera-Rueda CA, Stappenbeck T, Visperas A. Periprosthetic joint infection and immunity: Current understanding of host-microbe interplay. J Orthop Res 2024; 42:7-20. [PMID: 37874328 DOI: 10.1002/jor.25723] [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: 05/31/2023] [Revised: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Periprosthetic joint infection (PJI) is a major complication of total joint arthroplasty. Even with current treatments, failure rates are unacceptably high with a 5-year mortality rate of 26%. Majority of the literature in the field has focused on development of better biomarkers for diagnostics and treatment strategies including innovate antibiotic delivery systems, antibiofilm agents, and bacteriophages. Nevertheless, the role of the immune system, our first line of defense during PJI, is not well understood. Evidence of infection in PJI patients is found within circulation, synovial fluid, and tissue and include numerous cytokines, metabolites, antimicrobial peptides, and soluble receptors that are part of the PJI diagnosis workup. Macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs) are initially recruited into the joint by chemokines and cytokines produced by immune cells and bacteria and are activated by pathogen-associated molecular patterns. While these cells are efficient killers of planktonic bacteria by phagocytosis, opsonization, degranulation, and recruitment of adaptive immune cells, biofilm-associated bacteria are troublesome. Biofilm is not only a physical barrier for the immune system but also elicits effector functions. Additionally, bacteria have developed mechanisms to evade the immune system by inactivating effector molecules, promoting killing or anti-inflammatory effector cell phenotypes, and intracellular persistence and dissemination. Understanding these shortcomings and the mechanisms by which bacteria can subvert the immune system may open new approaches to better prepare our own immune system to combat PJI. Furthermore, preoperative immune system assessment and screening for dysregulation may aid in developing preventative interventions to decrease PJI incidence.
Collapse
Affiliation(s)
- Nicolas S Piuzzi
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alison K Klika
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| | - Qiuhe Lu
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Anabelle Visperas
- Department of Orthopaedic Surgery, Cleveland Clinic Adult Reconstruction Research (CCARR), Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
14
|
Arumugam P, Kielian T. Metabolism Shapes Immune Responses to Staphylococcus aureus. J Innate Immun 2023; 16:12-30. [PMID: 38016430 PMCID: PMC10766399 DOI: 10.1159/000535482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) is a common cause of hospital- and community-acquired infections that can result in various clinical manifestations ranging from mild to severe disease. The bacterium utilizes different combinations of virulence factors and biofilm formation to establish a successful infection, and the emergence of methicillin- and vancomycin-resistant strains introduces additional challenges for infection management and treatment. SUMMARY Metabolic programming of immune cells regulates the balance of energy requirements for activation and dictates pro- versus anti-inflammatory function. Recent investigations into metabolic adaptations of leukocytes and S. aureus during infection indicate that metabolic crosstalk plays a crucial role in pathogenesis. Furthermore, S. aureus can modify its metabolic profile to fit an array of niches for commensal or invasive growth. KEY MESSAGES Here we focus on the current understanding of immunometabolism during S. aureus infection and explore how metabolic crosstalk between the host and S. aureus influences disease outcome. We also discuss how key metabolic pathways influence leukocyte responses to other bacterial pathogens when information for S. aureus is not available. A better understanding of how S. aureus and leukocytes adapt their metabolic profiles in distinct tissue niches may reveal novel therapeutic targets to prevent or control invasive infections.
Collapse
Affiliation(s)
- Prabhakar Arumugam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
15
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
16
|
Tuek-Um S, Yangtara S, Surachetpong W, Kaewmongkol S, Kaewmongkol G, Thengchaisri N. Bacterial DNA and osteoarthritis in dogs with patellar luxation and cranial cruciate ligament rupture. Vet World 2023; 16:2049-2054. [PMID: 38023283 PMCID: PMC10668564 DOI: 10.14202/vetworld.2023.2049-2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/05/2023] [Indexed: 12/01/2023] Open
Abstract
Background and Aim The association between bacterial DNA in stifle joints, including those with cranial cruciate ligament rupture (CCLR) and medial patellar luxation (MPL), and osteoarthritis in dogs remains elusive. This study investigated the potential association between the detection of bacterial DNA and osteoarthritis in dogs using a broad-range polymerase chain reaction technique targeting the 16S ribosomal RNA gene. Materials and Methods Synovial fluid (35 samples) and knee tissue samples (32 samples) were obtained from 35 dogs diagnosed with CCLR (n = 20; 11 males and nine females) or MPL (n = 15; five males and 10 females) who underwent a surgical operation between October 2014 and April 2015. Results Dogs with CCLR had a higher average osteoarthritis score than those with MPL (2.0 ± 0.9 vs. 0.5 ± 0.9; p = 0.005). Bacterial DNA was detected in the stifle joints of 60.71% of dogs with MPL. Pelomonas spp. (25.00%), Halomonas spp. (17.86%), and 5 other species (17.86%) were the most frequently identified bacteria. Bacterial DNA was detected in 41.03% of dogs with CCLR. Pelomonas spp. (15.38%), Sphingomonas spp. (10.26%), Halomonas spp. (5.13%), and 4 other species (10.26%) were the most frequently identified bacteria. No significant difference was observed in the prevalence of bacterial DNA obtained from tissue samples (46.88%) or joint fluid samples (51.43%). The presence of bacterial DNA was not associated with the type of knee injury (MPL or CCLR; p = 1.000). There was a higher prevalence of bacterial DNA in samples from dogs with moderate-to-severe osteoarthritis (94.44%) than in those with minimal osteoarthritis (41.18%), and a significant association between the presence of bacterial DNA and moderate-to-severe osteoarthritis was identified (p < 0.01). Conclusion Dogs with moderate-to-severe osteoarthritis were more likely to have bacterial DNA in their stifle joints than those with no or minimal osteoarthritis. These findings provide valuable insight into the potential role of bacterial DNA in joint tissue or joint fluid and the development of osteoarthritis in dogs.
Collapse
Affiliation(s)
- Sirun Tuek-Um
- Surgery Unit, Kasetsart University Veterinary Teaching Hospital, Bangkhen campus, Bangkok 10900, Thailand
| | - Sarawut Yangtara
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| | - Win Surachetpong
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| | - Sarawan Kaewmongkol
- Department of Veterinary Nursing, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Gunn Kaewmongkol
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| | - Naris Thengchaisri
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
17
|
Goldmann O, Medina E. Myeloid-derived suppressor cells impair CD4+ T cell responses during chronic Staphylococcus aureus infection via lactate metabolism. Cell Mol Life Sci 2023; 80:221. [PMID: 37480485 PMCID: PMC10363054 DOI: 10.1007/s00018-023-04875-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 06/28/2023] [Accepted: 07/13/2023] [Indexed: 07/24/2023]
Abstract
Staphylococcus aureus is an important cause of chronic infections resulting from the failure of the host to eliminate the pathogen. Effective S. aureus clearance requires CD4+ T cell-mediated immunity. We previously showed that myeloid-derived suppressor cells (MDSC) expand during staphylococcal infections and support infection chronicity by inhibiting CD4+ T cell responses. The aim of this study was to elucidate the mechanisms underlying the suppressive effect exerted by MDSC on CD4+ T cells during chronic S. aureus infection. It is well known that activated CD4+ T cells undergo metabolic reprogramming from oxidative metabolism to aerobic glycolysis to meet their increased bioenergetic requirements. In this process, pyruvate is largely transformed into lactate by lactate dehydrogenase with the concomitant regeneration of NAD+, which is necessary for continued glycolysis. The by-product lactate needs to be excreted to maintain the glycolytic flux. Using SCENITH (single-cell energetic metabolism by profiling translation inhibition), we demonstrated here that MDSC inhibit CD4+ T cell responses by interfering with their metabolic activity. MDSC are highly glycolytic and excrete large amount of lactate in the local environment that alters the transmembrane concentration gradient and prevent removal of lactate by activated CD4+ T. Accumulation of endogenous lactate impedes the regeneration of NAD+, inhibit NAD-dependent glycolytic enzymes and stop glycolysis. Together, the results of this study have uncovered a role for metabolism on MDSC suppression of CD4+ T cell responses. Thus, reestablishment of their metabolic activity may represent a mean to improve the functionality of CD4+ T cells during chronic S. aureus infection.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.
| |
Collapse
|
18
|
Li M, Yu J, Guo G, Shen H. Interactions between Macrophages and Biofilm during Staphylococcus aureus-Associated Implant Infection: Difficulties and Solutions. J Innate Immun 2023; 15:499-515. [PMID: 37011602 PMCID: PMC10315156 DOI: 10.1159/000530385] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
Staphylococcus aureus (S. aureus) biofilm is the major cause of failure of implant infection treatment that results in heavy social and economic burden on individuals, families, and communities. Planktonic S. aureus attaches to medical implant surfaces where it proliferates and is wrapped by extracellular polymeric substances, forming a solid and complex biofilm. This provides a stable environment for bacterial growth, infection maintenance, and diffusion and protects the bacteria from antimicrobial agents and the immune system of the host. Macrophages are an important component of the innate immune system and resist pathogen invasion and infection through phagocytosis, antigen presentation, and cytokine secretion. The persistence, spread, or clearance of infection is determined by interplay between macrophages and S. aureus in the implant infection microenvironment. In this review, we discuss the interactions between S. aureus biofilm and macrophages, including the effects of biofilm-related bacteria on the macrophage immune response, roles of myeloid-derived suppressor cells during biofilm infection, regulation of immune cell metabolic patterns by the biofilm environment, and immune evasion strategies adopted by the biofilm against macrophages. Finally, we summarize the current methods that support macrophage-mediated removal of biofilms and emphasize the importance of considering multi-dimensions and factors related to implant-associated infection such as immunity, metabolism, the host, and the pathogen when developing new treatments.
Collapse
Affiliation(s)
- Mingzhang Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlong Yu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Geyong Guo
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Shen
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Maimaiti Z, Li Z, Xu C, Fu J, Hao LB, Chen JY, Chai W. Host Immune Regulation in Implant-Associated Infection (IAI): What Does the Current Evidence Provide Us to Prevent or Treat IAI? Bioengineering (Basel) 2023; 10:bioengineering10030356. [PMID: 36978747 PMCID: PMC10044746 DOI: 10.3390/bioengineering10030356] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The number of orthopedic implants for bone fixation and joint arthroplasty has been steadily increasing over the past few years. However, implant-associated infection (IAI), a major complication in orthopedic surgery, impacts the quality of life and causes a substantial economic burden on patients and societies. While research and study on IAI have received increasing attention in recent years, the failure rate of IAI has still not decreased significantly. This is related to microbial biofilms and their inherent antibiotic resistance, as well as the various mechanisms by which bacteria evade host immunity, resulting in difficulties in diagnosing and treating IAIs. Hence, a better understanding of the complex interactions between biofilms, implants, and host immunity is necessary to develop new strategies for preventing and controlling these infections. This review first discusses the challenges in diagnosing and treating IAI, followed by an extensive review of the direct effects of orthopedic implants, host immune function, pathogenic bacteria, and biofilms. Finally, several promising preventive or therapeutic alternatives are presented, with the hope of mitigating or eliminating the threat of antibiotic resistance and refractory biofilms in IAI.
Collapse
Affiliation(s)
- Zulipikaer Maimaiti
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhuo Li
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Chi Xu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jun Fu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Bo Hao
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Ji-Ying Chen
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- Correspondence: (J.-Y.C.); (W.C.)
| | - Wei Chai
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- Correspondence: (J.-Y.C.); (W.C.)
| |
Collapse
|
20
|
Tarabichi S, Chen AF, Higuera CA, Parvizi J, Polkowski GG. 2022 American Association of Hip and Knee Surgeons Symposium: Periprosthetic Joint Infection. J Arthroplasty 2023:S0883-5403(23)00065-7. [PMID: 36738863 DOI: 10.1016/j.arth.2023.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/06/2023] Open
Abstract
Periprosthetic joint infection (PJI) is the leading cause of failure in patients undergoing total joint arthroplasty. This article is a brief summary of a symposium on PJI that was presented at the annual AAHKS meeting. It will provide an overview of current technqiues in the prevention, diagnosis, and management of PJI. It will also highlight emerging technologies in this setting.
Collapse
Affiliation(s)
- Saad Tarabichi
- Rothman Orthopaedic Institute at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Antonia F Chen
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Carlos A Higuera
- Levitetz Department of Orthopaedic Surgery, Cleveland Clinic Florida, Weston, Florida
| | - Javad Parvizi
- Rothman Orthopaedic Institute at Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Gregory G Polkowski
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
21
|
Fisher CR, Patel R. Profiling the Immune Response to Periprosthetic Joint Infection and Non-Infectious Arthroplasty Failure. Antibiotics (Basel) 2023; 12:296. [PMID: 36830206 PMCID: PMC9951934 DOI: 10.3390/antibiotics12020296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Arthroplasty failure is a major complication of joint replacement surgery. It can be caused by periprosthetic joint infection (PJI) or non-infectious etiologies, and often requires surgical intervention and (in select scenarios) resection and reimplantation of implanted devices. Fast and accurate diagnosis of PJI and non-infectious arthroplasty failure (NIAF) is critical to direct medical and surgical treatment; differentiation of PJI from NIAF may, however, be unclear in some cases. Traditional culture, nucleic acid amplification tests, metagenomic, and metatranscriptomic techniques for microbial detection have had success in differentiating the two entities, although microbiologically negative apparent PJI remains a challenge. Single host biomarkers or, alternatively, more advanced immune response profiling-based approaches may be applied to differentiate PJI from NIAF, overcoming limitations of microbial-based detection methods and possibly, especially with newer approaches, augmenting them. In this review, current approaches to arthroplasty failure diagnosis are briefly overviewed, followed by a review of host-based approaches for differentiation of PJI from NIAF, including exciting futuristic combinational multi-omics methodologies that may both detect pathogens and assess biological responses, illuminating causes of arthroplasty failure.
Collapse
Affiliation(s)
- Cody R. Fisher
- Mayo Clinic Graduate School of Biomedical Sciences, Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Fisher CR, Krull JE, Bhagwate A, Masters T, Greenwood-Quaintance KE, Abdel MP, Patel R. Sonicate Fluid Cellularity Predicted by Transcriptomic Deconvolution Differentiates Infectious from Non-Infectious Arthroplasty Failure. J Bone Joint Surg Am 2023; 105:63-73. [PMID: 36574631 PMCID: PMC10137834 DOI: 10.2106/jbjs.22.00605] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Although cellularity is traditionally assessed morphologically, deep sequencing approaches being used for microorganism detection may be able to provide information about cellularity. We hypothesized that cellularity predicted using CIBERSORTx (Stanford University), a transcriptomic-based cellular deconvolution tool, would differentiate between infectious and non-infectious arthroplasty failure. METHODS CIBERSORTx-derived cellularity profiles of 93 sonicate fluid samples, including 53 from subjects who underwent failed arthroplasties due to periprosthetic joint infection (PJI) (abbreviated for the purpose of this study as PJIF) and 40 from subjects who had undergone non-infectious arthroplasty failure (abbreviated NIAF) that had been subjected to bulk RNA sequencing were evaluated. RESULTS Samples from PJIF and NIAF subjects were differentially clustered by principal component analysis based on the cellularity profile. Twelve of the 22 individual predicted cellular fractions were differentially expressed in the PJIF cases compared with the NIAF cases, including increased predicted neutrophils (mean and standard error, 9.73% ± 1.06% and 0.81% ± 0.60%), activated mast cells (17.12% ± 1.51% and 4.11% ± 0.44%), and eosinophils (1.96% ± 0.37% and 0.42% ± 0.21%), and decreased predicted M0 macrophages (21.33% ± 1.51% and 39.75% ± 2.45%), M2 macrophages (3.56% ± 0.52% and 8.70% ± 1.08%), and regulatory T cells (1.57% ± 0.23% and 3.20% ± 0.34%). The predicted total granulocyte fraction was elevated in the PJIF cases (32.97% ± 2.13% and 11.76% ± 1.61%), and the samples from the NIAF cases had elevated predicted total macrophage and monocyte (34.71% ± 1.71% and 55.34% ± 2.37%) and total B cell fractions (5.89% ± 0.30% and 8.62% ± 0.86%). Receiver operating characteristic curve analysis identified predicted total granulocytes, neutrophils, and activated mast cells as highly able to differentiate between the PJIF cases and the NIAF cases. Within the PJIF cases, the total granulocyte, total macrophage and monocyte, M0 macrophage, and M2 macrophage fractions were differentially expressed in Staphylococcus aureus compared with Staphylococcus epidermidis -associated samples. Within the NIAF cases, the predicted total B cell, naïve B cell, plasma cell, and M2 macrophage fractions were differentially expressed among different causes of failure. CONCLUSIONS CIBERSORTx can predict the cellularity of sonicate fluid using transcriptomic data, allowing for the evaluation of the underlying immune response during the PJIF and NIAF cases, without a need to phenotypically assess cell composition.
Collapse
Affiliation(s)
- Cody R Fisher
- Department of Immunology, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota.,Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jordan E Krull
- Department of Immunology, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota
| | - Aditya Bhagwate
- Department of Quantitative Sciences, Mayo Clinic, Rochester, Minnesota
| | - Thao Masters
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kerryl E Greenwood-Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.,Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
23
|
Ren Y, Bäcker H, Müller M, Kienzle A. The role of myeloid derived suppressor cells in musculoskeletal disorders. Front Immunol 2023; 14:1139683. [PMID: 36936946 PMCID: PMC10020351 DOI: 10.3389/fimmu.2023.1139683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The immune system is closely linked to bone homeostasis and plays a pivotal role in several pathological and inflammatory conditions. Through various pathways it modulates various bone cells and subsequently sustains the physiological bone metabolism. Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous immature myeloid-derived cells that can exert an immunosuppressive function through a direct cell-to-cell contact, secretion of anti-inflammatory cytokines or specific exosomes. These cells mediate the innate immune response to chronic stress on the skeletal system. In chronic inflammation, MDSCs act as an inner offset to rebalance overactivation of the immune system. Moreover, they have been found to be involved in processes responsible for bone remodeling in different musculoskeletal disorders, autoimmune diseases, infection, and cancer. These cells can not only cause bone erosion by differentiating into osteoclasts, but also alleviate the immune reaction, subsequently leading to long-lastingly impacted bone remodeling. In this review, we discuss the impact of MDSCs on the bone metabolism under several pathological conditions, the involved modulatory pathways as well as potential therapeutic targets in MDSCs to improve bone health.
Collapse
Affiliation(s)
- Yi Ren
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Henrik Bäcker
- Department of Orthopedics, Auckland City Hospital, Auckland, New Zealand
| | - Michael Müller
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Arne Kienzle
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité — Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Arne Kienzle,
| |
Collapse
|
24
|
Biedermann L, Bandick E, Ren Y, Tsitsilonis S, Donner S, Müller M, Duda G, Perka C, Kienzle A. Inflammation of Bone in Patients with Periprosthetic Joint Infections of the Knee. JB JS Open Access 2023; 8:e22.00101. [PMID: 36698987 PMCID: PMC9831161 DOI: 10.2106/jbjs.oa.22.00101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Despite the general success of total knee arthroplasty (TKA), addressing periprosthetic joint infection (PJI) and the resulting long-term complications is a growing medical need given the aging population and the increasing demand for arthroplasty. A larger proportion of patients face revision surgery because of the long-term complication of aseptic loosening despite clearance of the infection. The pathomechanisms leading to prosthetic loosening are not understood as it has been widely assumed that the bone stock recovers after explantation revision surgery. While clinical observations suggest a reduced osteogenic potential in patients with PJI, knowledge regarding the relevant biology is sparse. In the present study, we investigated the inflammatory impact of PJI on the bone and bone marrow in the vicinity of the joint. Additionally, we evaluated changes in the local inflammatory environment in a 2-stage exchange at both explantation and reimplantation. Methods In this study, we analyzed 75 human bone and bone-marrow specimens (obtained from 65 patients undergoing revision arthroplasty with cement for the treatment of PJI) for markers of inflammation. Samples were analyzed using hematoxylin and eosin overview staining, fluorescent immunohistochemical staining, flow cytometry, and polymerase chain reaction (PCR). Results Leukocyte prevalence was significantly elevated at explantation (femur, +218.9%; tibia, +134.2%). While leukocyte prevalence decreased at reimplantation (femur, -49.5%; tibia, -34.2%), the number of cells remained significantly higher compared with the control group (femur, +61.2%; tibia, +54.2%). Expression of inflammatory markers interleukin (IL)-1α (femur, +2,748.7%; tibia, +1,605.9%), IL-6 (femur, +2,062.5%; tibia, +2,385.7%), IL-10 (femur, +913.7%; tibia, +897.5%), IL-12 (femur, +386.1%; tibia, +52.5%), IL-18 (femur, +805.3%; tibia, +547.7%), and tumor necrosis factor (TNF)-α (femur, +296.9%; tibia, +220.9%) was significantly elevated at prosthesis explantation in both femoral and tibial specimens. Expression remained significantly elevated at reimplantation for all inflammatory markers except IL-12 compared with the control group. Conversely, there were only limited inflammatory changes in the bone marrow environment. Conclusions The present study demonstrated a strong and lasting upregulation of the proinflammatory environment in the joint-surrounding osseous scaffold in patients with PJI. Our data suggest that modulating the inflammatory environment has substantial potential to improve the clinical outcome in affected patients.
Collapse
Affiliation(s)
- Lara Biedermann
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Evgeniya Bandick
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Yi Ren
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Serafeim Tsitsilonis
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefanie Donner
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Michael Müller
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Georg Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Perka
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arne Kienzle
- Clinic for Orthopedics, Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
25
|
Warren SI, Charville GW, Manasherob R, Amanatullah DF. Immune checkpoint upregulation in periprosthetic joint infection. J Orthop Res 2022; 40:2663-2669. [PMID: 35124851 PMCID: PMC9352818 DOI: 10.1002/jor.25276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/06/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Periprosthetic joint infections (PJI) induce an immunosuppressive cytokine profile through an unknown mechanism. Immune checkpoints, like programmed cell death 1 (PD-1) and its ligand (PD-L1), initiate innate immunosuppressive pathways essential for self-tolerance. Several malignancies and chronic infections co-opt these pathways to derive a survival advantage. This study evaluates PD-1/PD-L1 expression in periprosthetic tissue from patients undergoing revision hip or knee arthroplasty for a PJI versus an aseptic failure. PD-1/PD-L1 in the global tissue sample and the high-power microscopic field of maximum expression was analyzed prospectively using immunohistochemistry. Fifteen patients with a PJI (45%) and 16 patients with an aseptic failure (52%) were included. PD-1 expression was uniformly low. Maximum PD-L1 expression was upregulated in patients with a PJI (25%, interquartile range [IQR]: 5%-75%) versus an aseptic failure, (8%, IQR: 1%-48%, p = 0.039). In the PJI cohort, maximum PD-L1 expression was higher among patients who developed a recurrent PJI (68%, IQR: 53%-86% vs. 15%, IQR: 5%-70%, p = 0.039). Patients with global PD-L1 over 5% trended toward a near 22-fold increase in the odds of reinfection (odds ratio [OR]: 21.9, 95% confidence interval [CI]: 0.9-523.5, p = 0.057) and patients with maximum PD-L1 over 20% trended toward a 15-fold increase in the odds of reinfection (OR: 15.0, 95% CI: 0.6-348.9, p = 0.092). These results support immune checkpoint upregulation as a mechanism of PJI-induced local immune dysfunction. Future studies should confirm PD-L1 as a risk factor for reinfection in larger cohorts.
Collapse
Affiliation(s)
- Shay I. Warren
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | | | - Robert Manasherob
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
26
|
Role of Staphylococcus aureus Formate Metabolism during Prosthetic Joint Infection. Infect Immun 2022; 90:e0042822. [PMID: 36286525 PMCID: PMC9670962 DOI: 10.1128/iai.00428-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Biofilms are bacterial communities characterized by antibiotic tolerance.
Staphylococcus aureus
is a leading cause of biofilm infections on medical devices, including prosthetic joints, which represent a significant health care burden. The major leukocyte infiltrate associated with
S. aureus
prosthetic joint infection (PJI) is granulocytic myeloid-derived suppressor cells (G-MDSCs), which produce IL-10 to promote biofilm persistence by inhibiting monocyte and macrophage proinflammatory activity.
Collapse
|
27
|
Sokhi UK, Xia Y, Sosa B, Turajane K, Nishtala SN, Pannellini T, Bostrom MP, Carli AV, Yang X, Ivashkiv LB. Immune Response to Persistent Staphyloccocus Aureus Periprosthetic Joint Infection in a Mouse Tibial Implant Model. J Bone Miner Res 2022; 37:577-594. [PMID: 34897801 PMCID: PMC8940655 DOI: 10.1002/jbmr.4489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/22/2021] [Accepted: 12/08/2021] [Indexed: 12/27/2022]
Abstract
Staphyloccocus aureus is one of the major pathogens in orthopedic periprosthetic joint infection (PJI), a devastating complication of total joint arthroplasty that often results in chronic and persistent infections that are refractory to antibiotics and require surgical interventions. Biofilm formation has been extensively investigated as a reason for persistent infection. The cellular composition, activation status, cytokine profile, and role of the immune response during persistent S. aureus PJI are incompletely understood. In this study, we used histology, multiparametric flow cytometry, and gene expression analysis to characterize the immune response in a clinically relevant orthopedic PJI model. We tested the hypothesis that persistent S. aureus infection induces feedback mechanisms that suppress immune cell activation, thereby affecting the course of infection. Surprisingly, persistent infection was characterized by strikingly high cytokine gene expression indicative of robust activation of multiple components of innate and adaptive immunity, along with ongoing severe neutrophil-dominated inflammation, in infected joint and bone tissues. Activation and expansion of draining lymph nodes and a bone marrow stress granulopoiesis reaction were also maintained during late phase infection. In parallel, feedback mechanisms involving T-cell inhibitory receptors and exhaustion markers, suppressive cytokines, and regulatory T cells were activated and associated with decreased T-cell proliferation and tissue infiltration during the persistent phase of infection. These results identify the cellular and molecular components of the mouse immune response to persistent S. aureus PJI and indicate that neutrophil infiltration, inflammatory cytokine responses, and ongoing lymph node and bone marrow reactions are insufficient to clear infection and that immune effector mechanisms are suppressed by feedback inhibitory pathways. These immune-suppressive mechanisms are associated with diminished T-cell proliferation and tissue infiltration and can be targeted as part of adjuvant immunotherapeutic strategies in combination with debridement of biofilm, antibiotics, and other therapeutic modalities to promote eradication of infection. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Upneet K Sokhi
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Yunwei Xia
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Branden Sosa
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Kathleen Turajane
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Sita N Nishtala
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Tania Pannellini
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Department of Pathology, Hospital for Special Surgery, New York, NY, USA
| | - Mathias P Bostrom
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA.,Department of Orthopaedics, Weill Cornell Medicine, New York, NY, USA
| | - Alberto V Carli
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,Arthroplasty Research Laboratory, Hospital for Special Surgery, New York, NY, USA
| | - Lionel B Ivashkiv
- Research Institute, Hospital for Special Surgery, New York, NY, USA.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.,Department of Medicine, Weill Cornell Medicine, New York, NY, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
28
|
Wang Y, Dikeman D, Zhang J, Ackerman N, Kim S, Alphonse MP, Ortines RV, Liu H, Joyce DP, Dillen CA, Thompson JM, Thomas AA, Plaut RD, Miller LS, Archer NK. CCR2 contributes to host defense against Staphylococcus aureus orthopedic implant-associated infections in mice. J Orthop Res 2022; 40:409-419. [PMID: 33713394 PMCID: PMC8435538 DOI: 10.1002/jor.25027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/19/2021] [Accepted: 03/10/2021] [Indexed: 02/04/2023]
Abstract
C-C motif chemokine receptor 2 (CCR2) is an important mediator of myeloid cell chemotaxis during inflammation and infection. Myeloid cells such as monocytes, macrophages, and neutrophils contribute to host defense during orthopedic implant-associated infections (OIAI), but whether CCR2-mediated chemotaxis is involved remains unclear. Therefore, a Staphylococcus aureus OIAI model was performed by surgically placing an orthopedic-grade titanium implant and inoculating a bioluminescent S. aureus strain in knee joints of wildtype (wt) and CCR2-deficient mice. In vivo bioluminescent signals significantly increased in CCR2-deficient mice compared with wt mice at later time points (Days 14-28), which was confirmed with ex vivo colony-forming unit enumeration. S. aureus γ-hemolysin utilizes CCR2 to induce host cell lysis. However, there were no differences in bacterial burden when the OIAI model was performed with a parental versus a mutant γ-hemolysin-deficient S. aureus strain, indicating that the protection was mediated by the host cell function of CCR2 rather than γ-hemolysin virulence. Although CCR2-deficient and wt mice had similar cellular infiltrates in the infected joint tissue, CCR2-deficient mice had reduced myeloid cells and γδ T cells in the draining lymph nodes. Taken together, CCR2 contributed to host defense at later time points during an OIAI by increasing immune cell infiltrates in the draining lymph nodes, which likely contained the infection and prevented invasive spread.
Collapse
Affiliation(s)
- Yu Wang
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Dustin Dikeman
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Nicole Ackerman
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Sophia Kim
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Roger V. Ortines
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - Carly A. Dillen
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| | - John M. Thompson
- Department of Orthopaedic Surgery, Johns Hopkins University
School of Medicine, Baltimore, Maryland, USA
| | - Abigail A. Thomas
- Division of Bacterial Parasitic, and Allergenic Products,
Center for Biologics Evaluation and Research, Food and Drug Administration, Silver
Spring, Maryland, USA
| | - Roger D. Plaut
- Division of Bacterial Parasitic, and Allergenic Products,
Center for Biologics Evaluation and Research, Food and Drug Administration, Silver
Spring, Maryland, USA
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA,Department of Immunology, Janssen Research and Development,
Spring House, Pennsylvania, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Mirzaei R, Sabokroo N, Ahmadyousefi Y, Motamedi H, Karampoor S. Immunometabolism in biofilm infection: lessons from cancer. Mol Med 2022; 28:10. [PMID: 35093033 PMCID: PMC8800364 DOI: 10.1186/s10020-022-00435-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Biofilm is a community of bacteria embedded in an extracellular matrix, which can colonize different human cells and tissues and subvert the host immune reactions by preventing immune detection and polarizing the immune reactions towards an anti-inflammatory state, promoting the persistence of biofilm-embedded bacteria in the host. MAIN BODY OF THE MANUSCRIPT It is now well established that the function of immune cells is ultimately mediated by cellular metabolism. The immune cells are stimulated to regulate their immune functions upon sensing danger signals. Recent studies have determined that immune cells often display distinct metabolic alterations that impair their immune responses when triggered. Such metabolic reprogramming and its physiological implications are well established in cancer situations. In bacterial infections, immuno-metabolic evaluations have primarily focused on macrophages and neutrophils in the planktonic growth mode. CONCLUSION Based on differences in inflammatory reactions of macrophages and neutrophils in planktonic- versus biofilm-associated bacterial infections, studies must also consider the metabolic functions of immune cells against biofilm infections. The profound characterization of the metabolic and immune cell reactions could offer exciting novel targets for antibiofilm therapy.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Niloofar Sabokroo
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Dietrich O, Heinz A, Goldmann O, Geffers R, Beineke A, Hiller K, Saliba AE, Medina E. Dysregulated Immunometabolism Is Associated with the Generation of Myeloid-Derived Suppressor Cells in Staphylococcus aureus Chronic Infection. J Innate Immun 2021; 14:257-274. [PMID: 34763332 DOI: 10.1159/000519306] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/23/2021] [Indexed: 11/19/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a compendium of immature myeloid cells that exhibit potent T-cell suppressive capacity and expand during pathological conditions such as cancer and chronic infections. Although well-characterized in cancer, the physiology of MDSCs in the infection setting remains enigmatic. Here, we integrated single-cell RNA sequencing (scRNA-seq) and functional metabolic profiling to gain deeper insights into the factors governing the generation and maintenance of MDSCs in chronic Staphylococcus aureus infection. We found that MDSCs originate not only in the bone marrow but also at extramedullary sites in S. aureus-infected mice. scRNA-seq showed that infection-driven MDSCs encompass a spectrum of myeloid precursors in different stages of differentiation, ranging from promyelocytes to mature neutrophils. Furthermore, the scRNA-seq analysis has also uncovered valuable phenotypic markers to distinguish mature myeloid cells from immature MDSCs. Metabolic profiling indicates that MDSCs exhibit high glycolytic activity and high glucose consumption rates, which are required for undergoing terminal maturation. However, rapid glucose consumption by MDSCs added to infection-induced perturbations in the glucose supplies in infected mice hinders the terminal maturation of MDSCs and promotes their accumulation in an immature stage. In a proof-of-concept in vivo experiment, we demonstrate the beneficial effect of increasing glucose availability in promoting MDSC terminal differentiation in infected mice. Our results provide valuable information of how metabolic alterations induced by infection influence reprogramming and differentiation of MDSCs.
Collapse
Affiliation(s)
- Oliver Dietrich
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Alexander Heinz
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany.,Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
31
|
In Vitro 3D Staphylococcus aureus Abscess Communities Induce Bone Marrow Cells to Expand into Myeloid-Derived Suppressor Cells. Pathogens 2021; 10:pathogens10111446. [PMID: 34832602 PMCID: PMC8622274 DOI: 10.3390/pathogens10111446] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 01/17/2023] Open
Abstract
Staphylococcus aureus is the main causative pathogen of subcutaneous, bone, and implant-related infections, forming structures known as staphylococcal abscess communities (SACs) within tissues that also contain immunosuppressive myeloid-derived suppressor cells (MDSCs). Although both SACs and MDSCs are present in chronic S. aureus infections, it remains unknown whether SACs directly trigger MDSC expansion. To investigate this, a previously developed 3D in vitro SAC model was co-cultured with murine and human bone marrow cells. Subsequently, it was shown that SAC-exposed human CD11blow/− myeloid cells or SAC-exposed murine CD11b+ Gr-1+ cells were immunosuppressive mainly by reducing absolute CD4+ and CD8α+ T cell numbers, as shown in T cell proliferation assays and with flow cytometry. Monocytic MDSCs from mice with an S. aureus bone infection also strongly reduced CD4+ and CD8α+ T cell numbers. Using protein biomarker analysis and an immunoassay, we detected in SAC–bone marrow co-cultures high levels of GM-CSF, IL-6, VEGF, IL-1β, TNFα, IL-10, and TGF-β. Furthermore, SAC-exposed neutrophils expressed Arg-1 and SAC-exposed monocytes expressed Arg-1 and iNOS, as shown via immunofluorescent stains. Overall, this study showed that SACs cause MDSC expansion from bone marrow cells and identified possible mediators to target as an additional strategy for treating chronic S. aureus infections.
Collapse
|
32
|
Veis DJ, Cassat JE. Infectious Osteomyelitis: Marrying Bone Biology and Microbiology to Shed New Light on a Persistent Clinical Challenge. J Bone Miner Res 2021; 36:636-643. [PMID: 33740314 DOI: 10.1002/jbmr.4279] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/01/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Infections of bone occur in a variety of clinical settings, ranging from spontaneous isolated infections arising from presumed hematogenous spread to those associated with skin and soft tissue wounds or medical implants. The majority are caused by the ubiquitous bacterium Staphyloccocus (S.) aureus, which can exist as a commensal organism on human skin as well as an invasive pathogen, but a multitude of other microbes are also capable of establishing bone infections. While studies of clinical isolates and small animal models have advanced our understanding of the role of various pathogen and host factors in infectious osteomyelitis (iOM), many questions remain unaddressed. Thus, there are many opportunities to elucidate host-pathogen interactions that may be leveraged toward treatment or prevention of this troublesome problem. Herein, we combine perspectives from bone biology and microbiology and suggest that interdisciplinary approaches will bring new insights to the field. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Deborah J Veis
- Division of Bone and Mineral Diseases, Departments of Medicine and Pathology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Shriners Hospitals for Children, St. Louis, MO, USA
| | - James E Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville,, TN, USA.,Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
33
|
de Morais SD, Kak G, Menousek JP, Kielian T. Immunopathogenesis of Craniotomy Infection and Niche-Specific Immune Responses to Biofilm. Front Immunol 2021; 12:625467. [PMID: 33708216 PMCID: PMC7940520 DOI: 10.3389/fimmu.2021.625467] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Bacterial infections in the central nervous system (CNS) can be life threatening and often impair neurological function. Biofilm infection is a complication following craniotomy, a neurosurgical procedure that involves the removal and replacement of a skull fragment (bone flap) to access the brain for surgical intervention. The incidence of infection following craniotomy ranges from 1% to 3% with approximately half caused by Staphylococcus aureus (S. aureus). These infections present a significant therapeutic challenge due to the antibiotic tolerance of biofilm and unique immune properties of the CNS. Previous studies have revealed a critical role for innate immune responses during S. aureus craniotomy infection. Experiments using knockout mouse models have highlighted the importance of the pattern recognition receptor Toll-like receptor 2 (TLR2) and its adaptor protein MyD88 for preventing S. aureus outgrowth during craniotomy biofilm infection. However, neither molecule affected bacterial burden in a mouse model of S. aureus brain abscess highlighting the distinctions between immune regulation of biofilm vs. planktonic infection in the CNS. Furthermore, the immune responses elicited during S. aureus craniotomy infection are distinct from biofilm infection in the periphery, emphasizing the critical role for niche-specific factors in dictating S. aureus biofilm-leukocyte crosstalk. In this review, we discuss the current knowledge concerning innate immunity to S. aureus craniotomy biofilm infection, compare this to S. aureus biofilm infection in the periphery, and discuss the importance of anatomical location in dictating how biofilm influences inflammatory responses and its impact on bacterial clearance.
Collapse
Affiliation(s)
- Sharon Db de Morais
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Gunjan Kak
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Joseph P Menousek
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, United States
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
34
|
Amin Yavari S, Castenmiller SM, van Strijp JAG, Croes M. Combating Implant Infections: Shifting Focus from Bacteria to Host. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002962. [PMID: 32914481 DOI: 10.1002/adma.202002962] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/28/2020] [Indexed: 05/06/2023]
Abstract
The widespread use of biomaterials to support or replace body parts is increasingly threatened by the risk of implant-associated infections. In the quest for finding novel anti-infective biomaterials, there generally has been a one-sided focus on biomaterials with direct antibacterial properties, which leads to excessive use of antibacterial agents, compromised host responses, and unpredictable effectiveness in vivo. This review sheds light on how host immunomodulation, rather than only targeting bacteria, can endow biomaterials with improved anti-infective properties. How antibacterial surface treatments are at risk to be undermined by biomaterial features that dysregulate the protection normally provided by critical immune cell subsets, namely, neutrophils and macrophages, is discussed. Accordingly, how the precise modification of biomaterial surface biophysical cues, or the incorporation of immunomodulatory drug delivery systems, can render biomaterials with the necessary immune-compatible and immune-protective properties to potentiate the host defense mechanisms is reviewed. Within this context, the protective role of host defense peptides, metallic particles, quorum sensing inhibitors, and therapeutic adjuvants is discussed. The highlighted immunomodulatory strategies may lay a foundation to develop anti-infective biomaterials, while mitigating the increasing threat of antibacterial drug resistance.
Collapse
Affiliation(s)
- Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Suzanne M Castenmiller
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, 3508GA, The Netherlands
| |
Collapse
|
35
|
High-Dimensional Analysis of Immune Cell Composition Predicts Periprosthetic Joint Infections and Dissects Its Pathophysiology. Biomedicines 2020; 8:biomedicines8090358. [PMID: 32957521 PMCID: PMC7554968 DOI: 10.3390/biomedicines8090358] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/06/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Accurate diagnosis of periprosthetic joint infections (PJI) is one of the most widely researched areas in modern orthopedic endoprosthesis. However, our understanding of the immunological basis of this severe complication is still limited. In this study, we developed a flow cytometric approach to precisely characterize the immune cell composition in periprosthetic joints. Using high-dimensional multi-parametric data, we defined, for the first time, the local immune cell populations of artificial joints. We identified significant differences in the cellular distribution between infected and non-infected samples, and revealed that myeloid-derived suppressor cells (MDSCs) act as potential regulators of infiltrating immune cells in PJI. Further, we developed an algorithm to predict septic and aseptic samples with high sensitivity and specificity, that may serve as an indispensable addition to the current criteria of the Musculoskeletal Infection Society. This study describes a novel approach to flow cytometrically analyze the immune cell infiltrate of joint fluid that not only improves our understanding of the pathophysiology of PJI, but also enables the development of a novel screening tool to predict infection status. Our data further suggest that pharmacological targeting of MDSCs represents a novel strategy for addressing PJI.
Collapse
|
36
|
Bosch ME, Bertrand BP, Heim CE, Alqarzaee AA, Chaudhari SS, Aldrich AL, Fey PD, Thomas VC, Kielian T. Staphylococcus aureus ATP Synthase Promotes Biofilm Persistence by Influencing Innate Immunity. mBio 2020; 11:e01581-20. [PMID: 32900803 PMCID: PMC7482063 DOI: 10.1128/mbio.01581-20] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/08/2020] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a major cause of prosthetic joint infection (PJI), which is characterized by biofilm formation. S. aureus biofilm skews the host immune response toward an anti-inflammatory profile by the increased recruitment of myeloid-derived suppressor cells (MDSCs) that attenuate macrophage proinflammatory activity, leading to chronic infection. A screen of the Nebraska Transposon Mutant Library identified several hits in the ATP synthase operon that elicited a heightened inflammatory response in macrophages and MDSCs, including atpA, which encodes the alpha subunit of ATP synthase. An atpA transposon mutant (ΔatpA) had altered growth kinetics under both planktonic and biofilm conditions, along with a diffuse biofilm architecture that was permissive for leukocyte infiltration, as observed by confocal laser scanning microscopy. Coculture of MDSCs and macrophages with ΔatpA biofilm elicited significant increases in the proinflammatory cytokines interleukin 12p70 (IL-12p70), tumor necrosis factor alpha (TNF-α), and IL-6. This was attributed to increased leukocyte survival resulting from less toxin and protease production by ΔatpA biofilm as determined by liquid chromatography with tandem mass spectrometry (LC-MS/MS). The enhanced inflammatory response elicited by ΔatpA biofilm was cell lysis-dependent since it was negated by polyanethole sodium sulfanate treatment or deletion of the major autolysin, Atl. In a mouse model of PJI, ΔatpA-infected mice had decreased MDSCs concomitant with increased monocyte/macrophage infiltrates and proinflammatory cytokine production, which resulted in biofilm clearance. These studies identify S. aureus ATP synthase as an important factor in influencing the immune response during biofilm-associated infection and bacterial persistence.IMPORTANCE Medical device-associated biofilm infections are a therapeutic challenge based on their antibiotic tolerance and ability to evade immune-mediated clearance. The virulence determinants responsible for bacterial biofilm to induce a maladaptive immune response remain largely unknown. This study identified a critical role for S. aureus ATP synthase in influencing the host immune response to biofilm infection. An S. aureus ATP synthase alpha subunit mutant (ΔatpA) elicited heightened proinflammatory cytokine production by leukocytes in vitro and in vivo, which coincided with improved biofilm clearance in a mouse model of prosthetic joint infection. The ability of S. aureus ΔatpA to augment host proinflammatory responses was cell lysis-dependent, as inhibition of bacterial lysis by polyanethole sodium sulfanate or a ΔatpAΔatl biofilm did not elicit heightened cytokine production. These studies reveal a critical role for AtpA in shaping the host immune response to S. aureus biofilm.
Collapse
Affiliation(s)
- Megan E Bosch
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Blake P Bertrand
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Abdulelah A Alqarzaee
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sujata S Chaudhari
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Amy L Aldrich
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Paul D Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vinai C Thomas
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
37
|
Trecourt A, Brevet M, Champagnac A, Conrad A, Josse J, Dupieux-Chabert C, Valour F, Ferry T. Plasma Cell Infiltration on Histopathological Samples of Chronic Bone and Joint Infections due to Cutibacterium acnes: A series of 21 Cases. J Bone Jt Infect 2020; 5:205-211. [PMID: 32670775 PMCID: PMC7358965 DOI: 10.7150/jbji.46187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/22/2020] [Indexed: 11/05/2022] Open
Abstract
Introduction: Histopathological definition of bone and joint infection (BJI) is based on Mirra's criterion (≥ 5 polymorphonuclears (PMNs) per field in 5 high power fields (HPFs)). However, this definition does not seem appropriate for chronic BJIs caused by slow-growing germs such as Cutibacterium acnes (C. acnes). The aim of this study was to confirm that Mirra's criterion is not adequate for diagnosis of BJIs due to C. acnes. The second objective was to determine if plasma cell infiltration could be useful for the diagnosis of chronic BJIs due to C. acnes. Methods: We retrospectively selected 25 consecutive patients from 2009 to 2013 with chronic BJIs due to C. acnes. Histological analysis was performed on the 21 cases with at least two C. acnes positive cultures. In addition of Mirra's criterion, the number of plasma cells (≥5 plasma cells/5 HPFs, defined as "CRIOAc Lyon's criterion") was implemented in the histopathological analysis. Patients were defined as infected, if at least one of the two criteria were present. Results: According to Mirra's and CRIOAc Lyon's criteria, positive histopathology was observed in 12 (57.1%) and 15 (71.4%) cases respectively. Considering the 9 cases with negative Mirra's criterion, high plasma cell infiltration (≥5 plasma cells per field/5 HPFs) was observed in 5 cases (55.6%), and low plasma cells infiltration (2-5 plasma cells per field/5 HPFs) was observed in 4 other cases (44.4%). Conclusions: Adding CRIOAc Lyon's criterion to Mirra's criterion might restore some histopathological diagnosis of chronic BJIs due to C. acnes when a chronic BJI is clinically suspected.
Collapse
Affiliation(s)
- Alexis Trecourt
- Hospices Civils de Lyon, Institut de pathologie multisites des Hospices Civils de Lyon, Site Est et plateforme de pathologie moléculaire, Bron, France
| | - Marie Brevet
- Hospices Civils de Lyon, Institut de pathologie multisites des Hospices Civils de Lyon, Site Est et plateforme de pathologie moléculaire, Bron, France.,Université Claude Bernard Lyon 1, Lyon, France
| | - Anne Champagnac
- Hospices Civils de Lyon, Institut de pathologie multisites des Hospices Civils de Lyon, Site Est et plateforme de pathologie moléculaire, Bron, France
| | - Anne Conrad
- Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Hôpital de la Croix-Rousse, Service de Maladies Infectieuses et Tropicales, Lyon, France.,Centre de Référence des Infections Ostéo- Articulaires Complexes de Lyon, Lyon, France.,Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| | - Jérôme Josse
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence des Infections Ostéo- Articulaires Complexes de Lyon, Lyon, France.,Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| | - Céline Dupieux-Chabert
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence des Infections Ostéo- Articulaires Complexes de Lyon, Lyon, France.,Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France.,Hospices Civils de Lyon, Hôpital Croix-Rousse, Institut des Agents Infectieux, Laboratoire de Bactériologie, Lyon, France
| | - Florent Valour
- Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Hôpital de la Croix-Rousse, Service de Maladies Infectieuses et Tropicales, Lyon, France.,Centre de Référence des Infections Ostéo- Articulaires Complexes de Lyon, Lyon, France.,Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| | - Tristan Ferry
- Université Claude Bernard Lyon 1, Lyon, France.,Hospices Civils de Lyon, Hôpital de la Croix-Rousse, Service de Maladies Infectieuses et Tropicales, Lyon, France.,Centre de Référence des Infections Ostéo- Articulaires Complexes de Lyon, Lyon, France.,Centre International de Recherche en Infectiologie, CIRI, Inserm U1111, CNRS UMR5308, ENS de Lyon, UCBL1, Lyon, France
| |
Collapse
|
38
|
Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogramme the host immune response during persistent infection. Nat Microbiol 2020; 5:1271-1284. [PMID: 32661313 PMCID: PMC7529909 DOI: 10.1038/s41564-020-0756-3] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 06/16/2020] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a leading cause of biofilm-associated prosthetic joint infection (PJI), resulting in significant disability and prolonged treatment. It is known that host leukocyte IL-10 production is required for S. aureus biofilm persistence in PJI. A S. aureus bursa aurealis Tn library consisting of 1,952 non-essential genes was screened for mutants that failed to induce IL-10 in myeloid-derived suppressor cells (MDSCs), which identified a critical role for bacterial lactic acid biosynthesis. We generated a S. aureus ddh/ldh1/ldh2 triple Tn mutant that cannot produce D- or L-lactate. Co-culture of MDSCs or macrophages with ddh/ldh1/ldh2 mutant biofilm produced substantially less IL-10 compared with wild type S. aureus, which was also observed in a mouse model of PJI and led to reduced biofilm burden. Using MDSCs recovered from the mouse PJI model and in vitro leukocyte-biofilm co-cultures we show that bacterial-derived lactate inhibits histone deacetylase 11 (HDAC11), causing unchecked HDAC6 activity and increased histone 3 acetylation at the Il-10 promoter, resulting in enhanced Il-10 transcription in MDSCs and macrophages. Finally, we show that synovial fluid of patients with PJI contains elevated amounts of D-lactate and IL-10 compared with control subjects, and bacterial lactate increases IL-10 production by human monocyte-derived macrophages. Biofilms are bacterial communities that are difficult to treat because of their tolerance to antibiotics and ability to evade immune-mediated clearance. Prosthetic joint infection (PJI), a devastating complication of arthroplasty, is characterized by biofilm formation. The current study has discovered a central role for lactic acid biosynthesis in S. aureus biofilm formation during PJI. Mechanistically, bacterial-derived lactate inhibits histone deacetylase 11 (HDAC11) activity, which causes extensive epigenetic changes at the promoters of numerous host genes, including the key anti-inflammatory cytokine Il-10. Indeed, IL-10 production by myeloid-derived suppressor cells (MDSCs) and macrophages is critical for biofilm persistence during PJI. HDAC11 inhibition by S. aureus lactate results in unchecked HDAC6 activity, a positive regulator of IL-10, thereby increasing IL-10 production by MDSCs and macrophages in vitro and in vivo. Similarly, S. aureus lactate promotes IL-10 production in human monocyte-derived macrophages following biofilm exposure. This study highlights how bacterial metabolism can influence the host immune response to promote infection persistence.
Collapse
|
39
|
Orthopaedic Surgery Elicits a Systemic Anti-Inflammatory Signature. J Clin Med 2020; 9:jcm9072123. [PMID: 32640676 PMCID: PMC7408679 DOI: 10.3390/jcm9072123] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022] Open
Abstract
Little information is available on the functional activity of leukocytes after arthroplasty or the expansion of populations with immune suppressive properties during the acute post-operative period. Synovial fluid and matched pre- and post-surgical blood samples were collected from total hip and knee arthroplasty patients (THA and TKA, respectively) to examine the impact of surgery on peripheral blood leukocyte frequency, bactericidal activity, and inflammatory mediator expression. For spinal surgeries, inflammatory mediator production by peripheral blood mononuclear cells (PBMCs) pre- and post-surgery was examined. An expansion of immune suppressive granulocytic myeloid-derived suppressor cells (G-MDSCs) was observed following arthroplasty, which correlated with significantly increased serum interleukin-10 (IL-10) levels. Analysis of synovial fluid from THA and TKAs revealed reduced granulocyte colony-stimulating factor (G-CSF) and soluble CD40 ligand (sCD40L) and increased interleukin-6 (IL-6), monocyte chemoattractant protein 2 (CCL2) and Fms-like tyrosine kinase 3 ligand (Flt-3L) compared to pre- and post-surgical serum. For the spinal surgery cohort, stimulation of PBMCs isolated post-surgery with bacterial antigens produced significantly less pro-inflammatory (IL-1α, IL-1β, interleukin-1 receptor antagonist (IL-1RA), IL-12p40, growth-related oncogene-α/GRO-α (CXCL1) and 6Ckine (CCL21)) and more anti-inflammatory/tissue repair mediators (IL-10, G-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF)) compared to PBMCs recovered before surgery. The observed bias towards systemic anti-inflammatory changes without concomitant increases in pro-inflammatory responses may influence susceptibility to infection following orthopaedic surgery in the context of underlying co-morbidities or risk factors.
Collapse
|
40
|
Yamada KJ, Heim CE, Xi X, Attri KS, Wang D, Zhang W, Singh PK, Bronich TK, Kielian T. Monocyte metabolic reprogramming promotes pro-inflammatory activity and Staphylococcus aureus biofilm clearance. PLoS Pathog 2020; 16:e1008354. [PMID: 32142554 PMCID: PMC7080272 DOI: 10.1371/journal.ppat.1008354] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 03/18/2020] [Accepted: 01/27/2020] [Indexed: 02/07/2023] Open
Abstract
Biofilm-associated prosthetic joint infections (PJIs) cause significant morbidity due to their recalcitrance to immune-mediated clearance and antibiotics, with Staphylococcus aureus (S. aureus) among the most prevalent pathogens. We previously demonstrated that S. aureus biofilm-associated monocytes are polarized to an anti-inflammatory phenotype and the adoptive transfer of pro-inflammatory macrophages attenuated biofilm burden, highlighting the critical role of monocyte/macrophage inflammatory status in dictating biofilm persistence. The inflammatory properties of leukocytes are linked to their metabolic state, and here we demonstrate that biofilm-associated monocytes exhibit a metabolic bias favoring oxidative phosphorylation (OxPhos) and less aerobic glycolysis to facilitate their anti-inflammatory activity and biofilm persistence. To shift monocyte metabolism in vivo and reprogram cells to a pro-inflammatory state, a nanoparticle approach was utilized to deliver the OxPhos inhibitor oligomycin to monocytes. Using a mouse model of S. aureus PJI, oligomycin nanoparticles were preferentially internalized by monocytes, which significantly reduced S. aureus biofilm burden by altering metabolism and promoting the pro-inflammatory properties of infiltrating monocytes as revealed by metabolomics and RT-qPCR, respectively. Injection of oligomycin alone had no effect on monocyte metabolism or biofilm burden, establishing that intracellular delivery of oligomycin is required to reprogram monocyte metabolic activity and that oligomycin lacks antibacterial activity against S. aureus biofilms. Remarkably, monocyte metabolic reprogramming with oligomycin nanoparticles was effective at clearing established biofilms in combination with systemic antibiotics. These findings suggest that metabolic reprogramming of biofilm-associated monocytes may represent a novel therapeutic approach for PJI.
Collapse
Affiliation(s)
- Kelsey J. Yamada
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Cortney E. Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Xinyuan Xi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Kuldeep S. Attri
- Eppley Institute, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Dezhen Wang
- Eppley Institute, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Wenting Zhang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Pankaj K. Singh
- Eppley Institute, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Tatiana K. Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
41
|
Campoccia D, Mirzaei R, Montanaro L, Arciola CR. Hijacking of immune defences by biofilms: a multifront strategy. BIOFOULING 2019; 35:1055-1074. [PMID: 31762334 DOI: 10.1080/08927014.2019.1689964] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 06/10/2023]
Abstract
Biofilm formation by pathogens and opportunistic bacteria is the basis of persistent or recurrent infections. Up to 80% of bacterial infections in humans are associated with biofilms. Despite the efficiency of the evolved and complex human defence system against planktonic bacteria, biofilms are capable of subverting host defences. The immune system is not completely effective in opposing bacteria and preventing infection. Increasing attention is being focussed on the mechanisms enabling bacterial biofilms to skew the coordinate action of humoral and cell mediated responses. Knowledge of the interactions between biofilm bacteria and the immune system is critical to effectively address biofilm infections, which have multiplied over the years with the spread of biomaterials in medicine. In this article, the latest information on the interactions between bacterial biofilms and immune cells is examined and the areas where of information is still lacking are explored.
Collapse
Affiliation(s)
- Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Rasoul Mirzaei
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Lucio Montanaro
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Carla Renata Arciola
- Laboratorio di Patologia delle Infezioni Associate all'Impianto, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
42
|
Seebach E, Kubatzky KF. Chronic Implant-Related Bone Infections-Can Immune Modulation be a Therapeutic Strategy? Front Immunol 2019; 10:1724. [PMID: 31396229 PMCID: PMC6664079 DOI: 10.3389/fimmu.2019.01724] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic implant-related bone infections are a major problem in orthopedic and trauma-related surgery with severe consequences for the affected patients. As antibiotic resistance increases in general and because most antibiotics have poor effectiveness against biofilm-embedded bacteria in particular, there is a need for alternative and innovative treatment approaches. Recently, the immune system has moved into focus as the key player in infection defense and bone homeostasis, and the targeted modulation of the host response is becoming an emerging field of interest. The aim of this review was to summarize the current knowledge of impaired endogenous defense mechanisms that are unable to prevent chronicity of bone infections associated with a prosthetic or osteosynthetic device. The presence of foreign material adversely affects the immune system by generating a local immune-compromised environment where spontaneous clearance of planktonic bacteria does not take place. Furthermore, the surface structure of the implant facilitates the transition of bacteria from the planktonic to the biofilm stage. Biofilm formation on the implant surface is closely linked to the development of a chronic infection, and a misled adaption of the immune system makes it impossible to effectively eliminate biofilm infections. The interaction between the immune system and bone cells, especially osteoclasts, is extensively studied in the field of osteoimmunology and this crosstalk further aggravates the course of bone infection by shifting bone homeostasis in favor of bone resorption. T cells play a major role in various chronic diseases and in this review a special focus was therefore set on what is known about an ineffective T cell response. Myeloid-derived suppressor cells (MDSCs), anti-inflammatory macrophages, regulatory T cells (Tregs) as well as osteoclasts all suppress immune defense mechanisms and negatively regulate T cell-mediated immunity. Thus, these cells are considered to be potential targets for immune therapy. The success of immune checkpoint inhibition in cancer treatment encourages the transfer of such immunological approaches into treatment strategies of other chronic diseases. Here, we discuss whether immune modulation can be a therapeutic tool for the treatment of chronic implant-related bone infections.
Collapse
Affiliation(s)
- Elisabeth Seebach
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| | - Katharina F Kubatzky
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
43
|
Josse J, Valour F, Maali Y, Diot A, Batailler C, Ferry T, Laurent F. Interaction Between Staphylococcal Biofilm and Bone: How Does the Presence of Biofilm Promote Prosthesis Loosening? Front Microbiol 2019; 10:1602. [PMID: 31379772 PMCID: PMC6653651 DOI: 10.3389/fmicb.2019.01602] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/26/2019] [Indexed: 12/19/2022] Open
Abstract
With the aging of population, the number of indications for total joint replacement is continuously increasing. However, prosthesis loosening can happen and is related to two major mechanisms: (1) aseptic loosening due to prosthesis micromotion and/or corrosion and release of wear particles from the different components of the implanted material and (2) septic loosening due to chronic prosthetic joint infection (PJI). The “aseptic” character of prosthesis loosening has been challenged over the years, especially considering that bacteria can persist in biofilms and be overlooked during diagnosis. Histological studies on periprosthetic tissue samples reported that macrophages are the principle cells associated with aseptic loosening due to wear debris. They produce cytokines and favor an inflammatory environment that induces formation and activation of osteoclasts, leading to bone resorption and periprosthetic osteolysis. In PJIs, the presence of infiltrates of polymorphonuclear neutrophils is a major criterion for histological diagnosis. Neutrophils are colocalized with osteoclasts and zones of osteolysis. A similar inflammatory environment also develops, leading to bone resorption through osteoclasts. Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis are the main staphylococci observed in PJIs. They share the common feature to form biofilm. For S. aureus and S. epidermidis, the interaction between biofilm and immunes cells (macrophages and polymorphonuclear neutrophils) differs regarding the species. Indeed, the composition of extracellular matrix of biofilm seems to impact the interaction with immune cells. Recent papers also reported the major role of myeloid-derived suppressor cells in biofilm-associated PJIs with S. aureus. These cells prevent lymphocyte infiltration and facilitate biofilm persistence. Moreover, the role of T lymphocytes is still unclear and potentially underestimates. In this review, after introducing the cellular mechanism of aseptic and septic loosening, we will focus on the interrelationships between staphylococcal biofilm, immune cells, and bone cells.
Collapse
Affiliation(s)
- Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Cécile Batailler
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Maladies Infectieuses, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Laboratoire de Bactériologie, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
44
|
Abstract
Staphylococci, with the leading species Staphylococcus aureus and Staphylococcus epidermidis, are the most frequent causes of infections on indwelling medical devices. The biofilm phenotype that those bacteria adopt during device-associated infection facilitates increased resistance to antibiotics and host immune defenses. This review presents and discusses the molecular mechanisms contributing to staphylococcal biofilm development and their in-vivo importance. Furthermore, it summarizes current strategies for the development of therapeutics against staphylococcal biofilm-associated infection.
Collapse
|
45
|
Peng KT, Chiang YC, Huang TY, Chen PC, Chang PJ, Lee CW. Curcumin nanoparticles are a promising anti-bacterial and anti-inflammatory agent for treating periprosthetic joint infections. Int J Nanomedicine 2019; 14:469-481. [PMID: 30666108 PMCID: PMC6333393 DOI: 10.2147/ijn.s191504] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Periprosthetic joint infections (PJIs) have a high incidence of recurrence after total joint replacement and are difficult to treat by debridement or antibiotic treatment. Curcumin is a natural product with anti-inflammatory and anti-bacterial properties. The low bioactivity of curcumin in water restricts its clinical application. Curcumin nanoparticles (CURN) were developed to overcome this limitation. Methods In this study, the therapeutic effects of CURN and their anti-inflammatory functions were investigated in a Staphylococcus aureus biofilm-induced PJIs model. Results CURN first attenuated the biofilm-induced expansion of myeloid-derived suppressor cells (MDSCs) and then regulated M1- and M2-phenotypic MDSC expression. Down-regulation of cytokines and reactive oxygen species was considered as the mechanism of CURN in reversing the suppression of T cell proliferation. The recovery of bone permeative destruction demonstrated that CURN enhanced therapeutic potency of vancomycin in vivo. Conclusion This is the first study to demonstrate that CURN may be useful for treating PJIs.
Collapse
Affiliation(s)
- Kuo-Ti Peng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan, .,College of Medicine, Chang Gung University, Guishan District, Taoyuan City 33303, Taiwan,
| | - Yao-Chang Chiang
- Department of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan, .,Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan,
| | - Tsung-Yu Huang
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Pei-Chun Chen
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan,
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang-Gung University, Taoyuan, Taiwan.,Department of Nephrology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chiang-Wen Lee
- Department of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan, .,Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan, .,Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Guishan District, Taoyuan City 33303, Taiwan, .,Department of Rehabilitation, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan,
| |
Collapse
|
46
|
Budhwar S, Verma P, Verma R, Rai S, Singh K. The Yin and Yang of Myeloid Derived Suppressor Cells. Front Immunol 2018; 9:2776. [PMID: 30555467 PMCID: PMC6280921 DOI: 10.3389/fimmu.2018.02776] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
In recent years, most of our knowledge about myeloid derived suppressor cells (MDSCs) has come from cancer studies, which depicts Yin side of MDSCs. In cancer, inherent immunosuppressive action of MDSCs favors tumor progression by inhibiting antitumor immune response. However, recently Yang side of MDSCs has also been worked out and suggests the role in maintenance of homeostasis during non-cancer situations like pregnancy, obesity, diabetes, and autoimmune disorders. Continued work in this area has armored the biological importance of these cells as master regulators of immune system and prompted scientists all over the world to look from a different perspective. Therefore, explicating Yin and Yang arms of MDSCs is obligatory to use it as a double edged sword in a much smarter way. This review is an attempt toward presenting a synergistic coalition of all the facts and controversies that exist in understanding MDSCs, bring them on the same platform and approach their "Yin and Yang" nature in a more comprehensive and coherent manner.
Collapse
Affiliation(s)
- Snehil Budhwar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Priyanka Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Rachna Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sangeeta Rai
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Kiran Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
47
|
Heterogeneity of Ly6G + Ly6C + Myeloid-Derived Suppressor Cell Infiltrates during Staphylococcus aureus Biofilm Infection. Infect Immun 2018; 86:IAI.00684-18. [PMID: 30249747 DOI: 10.1128/iai.00684-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 09/20/2018] [Indexed: 01/11/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature monocytes and granulocytes. While neutrophils (polymorphonuclear leukocytes [PMNs]) are classically identified as highly differentiated cells specialized for antimicrobial defense, our laboratory has reported minor contributions of PMNs to the immune response during Staphylococcus aureus biofilm infection. However, these two cell types can be difficult to differentiate because of shared surface marker expression. Here we describe a more refined approach to distinguish MDSCs from PMNs utilizing the integrin receptor CD11b combined with conventional Ly6G and Ly6C expression. This approach separated the Ly6G+ Ly6C+ population that we previously identified in a mouse model of S. aureus orthopedic implant infection into two subsets, namely, CD11bhigh Ly6G+ Ly6C+ MDSCs and CD11blow Ly6G+ Ly6C+ PMNs, which was confirmed by characteristic nuclear morphology using cytospins. CD11bhigh Ly6G+ Ly6C+ MDSCs suppressed T cell proliferation throughout the 28-day infection period, whereas CD11blow Ly6G+ Ly6C+ PMNs had no effect early (day 3 postinfection), although this population acquired suppressive activity at later stages of biofilm development. To further highlight the distinctions between biofilm-associated MDSCs and PMNs versus monocytes, transcriptional profiles were compared by transcriptome sequencing (RNA-Seq). A total of 6,466 genes were significantly differentially expressed in MDSCs versus monocytes, whereas only 297 genes were significantly different between MDSCs and PMNs. A number of genes implicated in cell cycle regulation were identified, and in vivo ethynyldeoxyuridine (EdU) labeling revealed that approximately 50% of MDSCs proliferated locally at the site of S. aureus biofilm infection. Based on their similar transcriptomic profiles to those of PMNs, biofilm-associated MDSCs are of a granulocytic lineage and can be classified as granulocytic MDSCs (G-MDSCs).
Collapse
|
48
|
Yamada KJ, Kielian T. Biofilm-Leukocyte Cross-Talk: Impact on Immune Polarization and Immunometabolism. J Innate Immun 2018; 11:280-288. [PMID: 30347401 DOI: 10.1159/000492680] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022] Open
Abstract
Biofilms are bacterial communities contained within an extracellular matrix, which can colonize both native tissues and artificial surfaces. In particular, indwelling medical devices and prosthetic implants are targets for biofilm formation because they facilitate bacterial attachment via host proteins that coat the foreign body. Biofilm infections are particularly challenging to treat, since they are not readily cleared by antibiotics, require invasive procedures to eradicate, and are prone to recurrence. It has been demonstrated that biofilm-derived products can actively suppress proinflammatory immune responses, as evident by the recruitment of myeloid-derived suppressor cells and macrophage (MФ) polarization towards an anti-inflammatory state. Recent studies have shown that alterations in leukocyte metabolism shape their inflammatory phenotype and function. For example, anti-inflammatory MФs are biased towards oxidative phosphorylation whereas proinflammatory MФs favor aerobic glycolysis. This review will compare the immune responses elicited by planktonic and biofilm bacterial infections, with a discussion on the metabolic properties of MФs and neutrophils in response to both bacterial growth conditions.
Collapse
Affiliation(s)
- Kelsey J Yamada
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA,
| |
Collapse
|