1
|
Van Roy Z, Kielian T. Immune-based strategies for the treatment of biofilm infections. Biofilm 2025; 9:100264. [PMID: 40093652 PMCID: PMC11909721 DOI: 10.1016/j.bioflm.2025.100264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Biofilms are bacterial communities surrounded by a polymeric matrix that can form on implanted materials and biotic surfaces, resulting in chronic infection that is recalcitrant to immune- and antibiotic-mediated clearance. Therefore, biofilm infections present a substantial clinical challenge, as treatment often involves additional surgical interventions to remove the biofilm nidus, prolonged antimicrobial therapy to clear residual bacteria, and considerable risk of treatment failure or infection recurrence. These factors, combined with progressive increases in antimicrobial resistance, highlight the need for alternative therapeutic strategies to circumvent undue morbidity, mortality, and resource strain on the healthcare system resulting from biofilm infections. One promising option is reprogramming dysfunctional immune responses elicited by biofilm. Here, we review the literature describing immune responses to biofilm infection with a focus on targets or strategies ripe for clinical translation. This represents a complex and dynamic challenge, with context-dependent host-pathogen interactions that differ across infection models, microenvironments, and individuals. Nevertheless, consistencies among these variables exist, which could facilitate the development of immune-based strategies for the future treatment of biofilm infections.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
2
|
Ogunware AE, Kielian T. Immunometabolism shapes chronic Staphylococcus aureus infection: insights from biofilm infection models. Curr Opin Microbiol 2025; 86:102612. [PMID: 40409167 DOI: 10.1016/j.mib.2025.102612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/17/2025] [Accepted: 04/28/2025] [Indexed: 05/25/2025]
Abstract
Staphylococcus aureus is both a commensal bacterium and versatile pathogen, capable of transitioning from a benign colonizer to cause invasive disease. Its ability to form biofilm - a resilient, highly structured bacterial community - plays a key role in chronic infections, including those associated with medical implants and native tissues. The unique microenvironments of these biofilm niches create challenges for the host immune system, complicating pathogen clearance. Immunometabolism, the interplay between immune function and metabolic programming, plays a crucial role in dictating how the host combats S. aureus biofilms. Leukocytes undergo profound metabolic changes in response to biofilm, which can lead to dysregulated immune responses and persistent infection. This review explores recent insights defining the metabolic landscape of immune responses to S. aureus biofilm with a focus on two clinically relevant models, namely, craniotomy and prosthetic joint infection.
Collapse
Affiliation(s)
- Adedayo E Ogunware
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
3
|
Kak G, Van Roy Z, Fallet RW, Korshoj LE, Kielian T. CD4+ T cell-innate immune crosstalk is critical during Staphylococcus aureus craniotomy infection. JCI Insight 2025; 10:e183327. [PMID: 39989461 PMCID: PMC11949042 DOI: 10.1172/jci.insight.183327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/08/2025] [Indexed: 02/25/2025] Open
Abstract
Access to the brain for treating neurological sequalae requires a craniotomy, which can be complicated by infection. Staphylococcus aureus accounts for half of craniotomy infections, increasing morbidity in a medically fragile patient population. T cells preferentially traffic to the brain during craniotomy infection; however, their functional importance is unknown. Using a mouse model of S. aureus craniotomy infection, CD4+ T cells were critical for bacterial containment, as treatment of WT animals with anti-CD4 exacerbated infection that was similar to phenotypes in Rag1-/- mice. Single-cell RNA-Seq (scRNA-Seq) revealed transcriptional heterogeneity in brain CD3+ infiltrates, with CD4+ cells most prominent that displayed Th1- and Th17-like characteristics, and adoptive transfer of either subset in Rag1-/- animals during early infection prevented S. aureus outgrowth. scRNA-Seq identified a robust IFN signature in several innate immune clusters, and examination of cell-to-cell interactions revealed extensive T cell crosstalk with monocytes/macrophages that was also observed in human craniotomy infection. A cooperative role for Th1 and Th17 responses was demonstrated by treatment of Ifng-/- mice with IL-17A neutralizing antibody that recapitulated phenotypes in Rag1-/- animals. Collectively, these findings implicate Th1- and Th17-mediated proinflammatory responses in shaping the innate immune landscape for S. aureus containment during craniotomy infection.
Collapse
|
4
|
Van Roy Z, Kak G, Fallet RW, Kielian T. Interferon-gamma receptor signaling regulates innate immunity during Staphylococcus aureus craniotomy infection. J Neuroinflammation 2025; 22:46. [PMID: 39987156 PMCID: PMC11847343 DOI: 10.1186/s12974-025-03376-9] [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: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 02/24/2025] Open
Abstract
A craniotomy is a neurosurgical procedure performed to access the intracranial space. In 3-5% of cases, infections can develop, most caused by Staphylococcus aureus biofilm formation on the skull surface. Medical management of this infection is difficult, as biofilm properties confer immune and antimicrobial recalcitrance to the infection and necessitate additional surgical procedures. Furthermore, treatment failure rates can be appreciably high. These factors, compounded with rapidly expanding rates of antimicrobial resistance, highlight the need to develop alternative treatment strategies to target and reverse the immune dysfunction that occurs during biofilm infection. Our recent work has identified CD4+ Th1 and Th17 cells as potent regulators of innate immune cell activation during craniotomy infection. Here, we report the role of IFN-γ, versus other Th1- and Th17-derived cytokines, in programing the immune response to biofilm infection using both global and cell type-specific IFN-γR1-deficient (Ifngr1-/-) mice. Bacterial burdens were significantly higher in Ifngr1-/- relative to WT animals despite few changes in immune cell abundance. Single-cell transcriptomics identified candidate explanations for this phenotype as alterations in cell death pathways, innate immune cell activation, MHC-II expression, and T cell responses were significantly reduced in Ifngr1-/- mice. While caspase-1 activation in PMNs and macrophage/microglial MHC-II expression were regulated by IFN-γ signaling, no phenotypes were observed with either granulocyte- or macrophage/microglia Ifngr1-/- conditional knockout mice, suggestive of redundancy. Instead, a decreased Th1/Th17 ratio was identified in Ifngr1-/- animals that was corroborated by elevated IL-17 levels and correlated with dysfunctional T cell-innate immune communication. Further, Th17 cells were less effective than Th1 cells in promoting S. aureus bactericidal activity in microglia and macrophages. Collectively, this work identifies a key protective role for IFN-γ during craniotomy infection by enhancing macrophage and microglial antibacterial activity. Therefore, controlled programming of IFN-γ responses may represent a novel therapeutic strategy for chronic craniotomy infections.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Gunjan Kak
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Rachel W Fallet
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, Nebraska, 68198, USA.
| |
Collapse
|
5
|
Van Roy Z, Kak G, Korshoj LE, Menousek JP, Heim CE, Fallet RW, Campbell JR, Geary CR, Liu B, Gorantla S, Poluektova LY, Duan B, Campbell WS, Thorell WE, Kielian T. Single-cell profiling reveals a conserved role for hypoxia-inducible factor signaling during human craniotomy infection. Cell Rep Med 2024; 5:101790. [PMID: 39426374 PMCID: PMC11604514 DOI: 10.1016/j.xcrm.2024.101790] [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: 12/27/2023] [Revised: 05/16/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
Neurosurgeries complicated by infection are associated with prolonged treatment and significant morbidity. Craniotomy is a common neurosurgical procedure; however, the cellular and molecular signatures associated with craniotomy infection in human subjects are unknown. A retrospective study of over 2,500 craniotomies reveals diverse patient demographics, pathogen identity, and surgical landscapes associated with infection. Leukocyte profiling in patient tissues from craniotomy infection characterizes a predominance of granulocytic myeloid-derived suppressor cells that may arise from transmigrated blood neutrophils, based on single-cell RNA sequencing (scRNA-seq) trajectory analysis. Single-cell transcriptomic analysis identifies metabolic shifts in tissue leukocytes, including a conserved hypoxia-inducible factor (HIF) signature. The importance of HIF signaling was validated using a mouse model of Staphylococcus aureus craniotomy infection, where HIF inhibition increases chemokine production and leukocyte recruitment, exacerbating tissue pathology. These findings establish conserved metabolic and transcriptional signatures that may represent promising future therapeutic targets for human craniotomy infection in the face of increasing antimicrobial resistance.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gunjan Kak
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lee E Korshoj
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Joseph P Menousek
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cortney E Heim
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rachel W Fallet
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - James R Campbell
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Carol R Geary
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bo Liu
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - W Scott Campbell
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - William E Thorell
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
6
|
Karyagina AS, Grishin AV, Kudinova AG, Bulygina IN, Koudan EV, Orlova PA, Datsenko VP, Zhulina AV, Grunina TM, Poponova MS, Krivozubov MS, Gromova MS, Strukova NV, Generalova MS, Nikitin KE, Shchetinin IV, Luchnikov LO, Zaitseva SV, Kirsanova MA, Statnik ES, Senatov FS, Lunin VG, Gromov AV. Dual-Functional Implant Based on Gellan-Xanthan Hydrogel with Diopside, BMP-2 and Lysostaphin for Bone Defect Repair and Control of Staphylococcal Infection. Macromol Biosci 2024; 24:e2400205. [PMID: 39140453 DOI: 10.1002/mabi.202400205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/10/2024] [Indexed: 08/15/2024]
Abstract
A new dual-functional implant based on gellan-xanthan hydrogel with calcium-magnesium silicate ceramic diopside and recombinant lysostaphin and bone morphogenetic protein 2 (BMP-2)-ray is developed. In this composite, BMP-2 is immobilized on microparticles of diopside while lysostaphin is mixed directly into the hydrogel, providing sustained release of BMP-2 to allow gradual bone formation and rapid release of lysostaphin to eliminate infection immediately after implantation. Introduction of diopside of up to 3% (w/v) has a negligible effect on the mechanical properties of the hydrogel but provides a high sorption capacity for BMP-2. The hydrogels show good biocompatibility and antibacterial activity. Lysostaphin released from the implants over a 3 h period efficiently kills planktonic cells and completely destroys 24 h pre-formed biofilms of Staphylococcus aureus. Furthermore, in vivo experiments in a mouse model of critically-sized cranial defects infected with S. aureus show a complete lack of osteogenesis when implants contain only BMP-2, whereas, in the presence of lysostaphin, complete closure of the defect with newly formed mineralized bone tissue is observed. Thus, the new implantable gellan-xanthan hydrogel with diopside and recombinant lysostaphin and BMP-2 shows both osteogenic and antibacterial properties and represents a promising material for the treatment and/or prevention of osteomyelitis after bone trauma.
Collapse
Affiliation(s)
- Anna S Karyagina
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- All-Russia Research Institute of Agricultural Biotechnology, Russian Academy of Sciences, Moscow, 127550, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia
- Institute of Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Alexander V Grishin
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- All-Russia Research Institute of Agricultural Biotechnology, Russian Academy of Sciences, Moscow, 127550, Russia
| | - Alina G Kudinova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Inna N Bulygina
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- Institute of Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Elizaveta V Koudan
- Institute of Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Polina A Orlova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Vera P Datsenko
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Anna V Zhulina
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Tatyana M Grunina
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- All-Russia Research Institute of Agricultural Biotechnology, Russian Academy of Sciences, Moscow, 127550, Russia
| | - Maria S Poponova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Mikhail S Krivozubov
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Maria S Gromova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Natalia V Strukova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Maria S Generalova
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Kirill E Nikitin
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| | - Igor V Shchetinin
- Material Science Department, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Lev O Luchnikov
- LASE - Laboratory of Advanced Solar Energy, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Svetlana V Zaitseva
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- Institute of Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | | | - Eugene S Statnik
- "LUCh" Laboratory, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Fedor S Senatov
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- Institute of Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, 119049, Russia
| | - Vladimir G Lunin
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
- All-Russia Research Institute of Agricultural Biotechnology, Russian Academy of Sciences, Moscow, 127550, Russia
| | - Alexander V Gromov
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, Moscow, 123098, Russia
| |
Collapse
|
7
|
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
|
8
|
Van Roy Z, Shi W, Kak G, Duan B, Kielian T. Epigenetic Regulation of Leukocyte Inflammatory Mediator Production Dictates Staphylococcus aureus Craniotomy Infection Outcome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:414-428. [PMID: 37314520 PMCID: PMC10524781 DOI: 10.4049/jimmunol.2300050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023]
Abstract
Staphylococcus aureus is a common cause of surgical-site infections, including those arising after craniotomy, which is performed to access the brain for the treatment of tumors, epilepsy, or hemorrhage. Craniotomy infection is characterized by complex spatial and temporal dynamics of leukocyte recruitment and microglial activation. We recently identified unique transcriptional profiles of these immune populations during S. aureus craniotomy infection. Epigenetic processes allow rapid and reversible control over gene transcription; however, little is known about how epigenetic pathways influence immunity to live S. aureus. An epigenetic compound library screen identified bromodomain and extraterminal domain-containing (BET) proteins and histone deacetylases (HDACs) as critical for regulating TNF, IL-6, IL-10, and CCL2 production by primary mouse microglia, macrophages, neutrophils, and granulocytic myeloid-derived suppressor cells in response to live S. aureus. Class I HDACs (c1HDACs) were increased in these cell types in vitro and in vivo during acute disease in a mouse model of S. aureus craniotomy infection. However, substantial reductions in c1HDACs were observed during chronic infection, highlighting temporal regulation and the importance of the tissue microenvironment for dictating c1HDAC expression. Microparticle delivery of HDAC and BET inhibitors in vivo caused widespread decreases in inflammatory mediator production, which significantly increased bacterial burden in the brain, galea, and bone flap. These findings identify histone acetylation as an important mechanism for regulating cytokine and chemokine production across diverse immune cell lineages that is critical for bacterial containment. Accordingly, aberrant epigenetic regulation may be important for promoting S. aureus persistence during craniotomy infection.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Gunjan Kak
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
9
|
Kak G, Van Roy Z, Heim CE, Fallet RW, Shi W, Roers A, Duan B, Kielian T. IL-10 production by granulocytes promotes Staphylococcus aureus craniotomy infection. J Neuroinflammation 2023; 20:114. [PMID: 37179295 PMCID: PMC10183138 DOI: 10.1186/s12974-023-02798-7] [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] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Treatment of brain tumors, epilepsy, or hemodynamic abnormalities requires a craniotomy to access the brain. Nearly 1 million craniotomies are performed in the US annually, which increase to ~ 14 million worldwide and despite prophylaxis, infectious complications after craniotomy range from 1 to 3%. Approximately half are caused by Staphylococcus aureus (S. aureus), which forms a biofilm on the bone flap that is recalcitrant to antibiotics and immune-mediated clearance. However, the mechanisms responsible for the persistence of craniotomy infection remain largely unknown. The current study examined the role of IL-10 in promoting bacterial survival. METHODS A mouse model of S. aureus craniotomy infection was used with wild type (WT), IL-10 knockout (KO), and IL-10 conditional KO mice where IL-10 was absent in microglia and monocytes/macrophages (CX3CR1CreIL-10 fl/fl) or neutrophils and granulocytic myeloid-derived suppressor cells (G-MDSCs; Mrp8CreIL-10 fl/fl), the major immune cell populations in the infected brain vs. subcutaneous galea, respectively. Mice were examined at various intervals post-infection to quantify bacterial burden, leukocyte recruitment, and inflammatory mediator production in the brain and galea to assess the role of IL-10 in craniotomy persistence. In addition, the role of G-MDSC-derived IL-10 on neutrophil activity was examined. RESULTS Granulocytes (neutrophils and G-MDSCs) were the major producers of IL-10 during craniotomy infection. Bacterial burden was significantly reduced in IL-10 KO mice in the brain and galea at day 14 post-infection compared to WT animals, concomitant with increased CD4+ and γδ T cell recruitment and cytokine/chemokine production, indicative of a heightened proinflammatory response. S. aureus burden was reduced in Mrp8CreIL-10 fl/fl but not CX3CR1CreIL-10 fl/fl mice that was reversed following treatment with exogenous IL-10, suggesting that granulocyte-derived IL-10 was important for promoting S. aureus craniotomy infection. This was likely due, in part, to IL-10 production by G-MDSCs that inhibited neutrophil bactericidal activity and TNF production. CONCLUSION Collectively, these findings reveal a novel role for granulocyte-derived IL-10 in suppressing S. aureus clearance during craniotomy infection, which is one mechanism to account for biofilm persistence.
Collapse
Affiliation(s)
- Gunjan Kak
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Rachel W Fallet
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Wen Shi
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Axel Roers
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
10
|
Menousek J, Horn CM, Heim CE, Van Roy Z, Korshoj LE, Kielian T. Transcriptional Profiling of Phagocytic Leukocytes and Microglia Reveals a Critical Role for Reactive Oxygen Species in Biofilm Containment during Staphylococcus aureus Craniotomy Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1973-1986. [PMID: 36426943 PMCID: PMC9643635 DOI: 10.4049/jimmunol.2200503] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/13/2022] [Indexed: 12/31/2022]
Abstract
Craniotomies are performed to treat a variety of intracranial pathology. Surgical site infection remains a complication of craniotomy despite the use of prophylactic antibiotics and universal sterile precautions. Infections occur in 1-3% of procedures, with approximately half caused by Staphylococcus aureus that forms a biofilm on the bone flap and is recalcitrant to systemic antibiotic therapy. We used an S. aureus-dsRed construct to compare the phagocytic capacity of leukocytes and microglia in vitro and in vivo using a mouse model of craniotomy infection. In addition, single-cell RNA sequencing (scRNA-seq) was applied to determine whether a transcriptional signature could be identified for phagocytic versus nonphagocytic cells in vivo. S. aureus was phagocytosed to equivalent extents in microglia, macrophages, neutrophils, and granulocytic myeloid-derived suppressor cells in vitro; however, microglial uptake of S. aureus was limited in vivo, whereas the other leukocyte populations exhibited phagocytic activity. scRNA-seq comparing the transcriptional signatures of phagocytic (S. aureus-dsRed+) versus nonphagocytic (S. aureus-dsRed-) leukocytes identified classical pathways enriched in phagocytic cells (i.e., reactive oxygen species [ROS]/reactive nitrogen species, lysosome, iron uptake, and transport), whereas nonphagocytic populations had increased ribosomal, IFN, and hypoxia signatures. scRNA-seq also revealed a robust ROS profile, which led to the exploration of craniotomy infection in NADPH oxidase 2 knockout mice. S. aureus burden, leukocyte recruitment, and intracellular bacterial load were significantly increased in NADPH oxidase 2 KO compared with wild-type animals. Collectively, these results highlight the importance of ROS generation in phagocytes for S. aureus biofilm containment, but not clearance, during craniotomy infection.
Collapse
Affiliation(s)
- Joseph Menousek
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE 68198
| | - Christopher M. Horn
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Cortney E. Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Lee E. Korshoj
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
11
|
Korshoj LE, Shi W, Duan B, Kielian T. The Prospect of Nanoparticle Systems for Modulating Immune Cell Polarization During Central Nervous System Infection. Front Immunol 2021; 12:670931. [PMID: 34248952 PMCID: PMC8260670 DOI: 10.3389/fimmu.2021.670931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/03/2021] [Indexed: 01/20/2023] Open
Abstract
The blood-brain barrier (BBB) selectively restricts the entry of molecules from peripheral circulation into the central nervous system (CNS) parenchyma. Despite this protective barrier, bacteria and other pathogens can still invade the CNS, often as a consequence of immune deficiencies or complications following neurosurgical procedures. These infections are difficult to treat since many bacteria, such as Staphylococcus aureus, encode a repertoire of virulence factors, can acquire antibiotic resistance, and form biofilm. Additionally, pathogens can leverage virulence factor production to polarize host immune cells towards an anti-inflammatory phenotype, leading to chronic infection. The difficulty of pathogen clearance is magnified by the fact that antibiotics and other treatments cannot easily penetrate the BBB, which requires extended regimens to achieve therapeutic concentrations. Nanoparticle systems are rapidly emerging as a promising platform to treat a range of CNS disorders. Nanoparticles have several advantages, as they can be engineered to cross the BBB with specific functionality to increase cellular and molecular targeting, have controlled release of therapeutic agents, and superior bioavailability and circulation compared to traditional therapies. Within the CNS environment, therapeutic actions are not limited to directly targeting the pathogen, but can also be tailored to modulate immune cell activation to promote infection resolution. This perspective highlights the factors leading to infection persistence in the CNS and discusses how novel nanoparticle therapies can be engineered to provide enhanced treatment, specifically through modulation of immune cell polarization.
Collapse
Affiliation(s)
- Lee E. Korshoj
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, 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
|
12
|
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: 23] [Impact Index Per Article: 5.8] [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
|
13
|
Aldrich AL, Horn CM, Heim CE, Korshoj LE, Kielian T. Transcriptional Diversity and Niche-Specific Distribution of Leukocyte Populations during Staphylococcus aureus Craniotomy-Associated Biofilm Infection. THE JOURNAL OF IMMUNOLOGY 2021; 206:751-765. [PMID: 33419769 DOI: 10.4049/jimmunol.2001042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/06/2020] [Indexed: 12/17/2022]
Abstract
Neurosurgery for brain tumor resection or epilepsy treatment requires a craniotomy to gain access to the brain. Despite prophylactic measures, infectious complications occur at a frequency of 1-3%, with approximately half caused by Staphylococcus aureus (S. aureus) that forms a biofilm on the bone flap and is recalcitrant to antibiotics. Using single-cell RNA sequencing in a mouse model of S. aureus craniotomy infection, this study revealed the complex transcriptional heterogeneity of resident microglia and infiltrating monocytes in the brain, in addition to transcriptionally diverse granulocyte subsets in the s.c. galea and bone flap. In the brain, trajectory analysis identified the transition of microglia from a homeostatic/anti-inflammatory to proinflammatory and proliferative populations, whereas granulocytes in the brain demonstrated a trajectory from a granulocyte myeloid-derived suppressor cell (MDSC)-like phenotype to a small population of mature polymorphonuclear neutrophils (PMNs). In the galea, trajectory analysis identified the progression from two distinct granulocyte-MDSC-like populations to PMN clusters enriched for IFN signaling and cell cycle genes. Based on their abundance in the galea and bone flap, PMNs and MDSCs were depleted using anti-Ly6G, which resulted in increased bacterial burden. This revealed a critical role for PMNs in S. aureus containment because MDSCs were found to attenuate PMN antibacterial activity, which may explain, in part, why craniotomy infection persists in the presence of PMN infiltrates. These results demonstrate the existence of a transcriptionally diverse leukocyte response that likely influences the chronicity of S. aureus craniotomy infection.
Collapse
Affiliation(s)
- Amy L Aldrich
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Christopher M Horn
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Lee E Korshoj
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
14
|
Aldrich AL, Heim CE, Shi W, Fallet RW, Duan B, Kielian T. TLR2 and caspase-1 signaling are critical for bacterial containment but not clearance during craniotomy-associated biofilm infection. J Neuroinflammation 2020; 17:114. [PMID: 32290861 PMCID: PMC7158029 DOI: 10.1186/s12974-020-01793-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A craniotomy is required to access the brain for tumor resection or epilepsy treatment, and despite precautionary measures, infectious complications occur at a frequency of 1-3%. Approximately half of craniotomy infections are caused by Staphylococcus aureus (S. aureus) that forms a biofilm on the bone flap, which is recalcitrant to antibiotics. Our prior work in a mouse model of S. aureus craniotomy infection revealed a critical role for myeloid differentiation factor 88 (MyD88) in bacterial containment and pro-inflammatory mediator production. Since numerous receptors utilize MyD88 as a signaling adaptor, the current study examined the importance of Toll-like receptor 2 (TLR2) and TLR9 based on their ability sense S. aureus ligands, namely lipoproteins and CpG DNA motifs, respectively. We also examined the role of caspase-1 based on its known association with TLR signaling to promote IL-1β release. METHODS A mouse model of craniotomy-associated biofilm infection was used to investigate the role of TLR2, TLR9, and caspase-1 in disease progression. Wild type (WT), TLR2 knockout (KO), TLR9 KO, and caspase-1 KO mice were examined at various intervals post-infection to quantify bacterial burden, leukocyte recruitment, and inflammatory mediator production in the galea, brain, and bone flap. In addition, the role of TLR2-dependent signaling during microglial/macrophage crosstalk with myeloid-derived suppressor cells (MDSCs) was examined. RESULTS TLR2, but not TLR9, was important for preventing S. aureus outgrowth during craniotomy infection, as revealed by the elevated bacterial burden in the brain, galea, and bone flap of TLR2 KO mice concomitant with global reductions in pro-inflammatory mediator production compared to WT animals. Co-culture of MDSCs with microglia or macrophages, to model interactions in the brain vs. galea, respectively, also revealed a critical role for TLR2 in triggering pro-inflammatory mediator production. Similar to TLR2, caspase-1 KO animals also displayed increased S. aureus titers coincident with reduced pro-inflammatory mediator release, suggestive of pathway cooperativity. Treatment of caspase-1 KO mice with IL-1β microparticles significantly reduced S. aureus burden in the brain and galea compared to empty microparticles, confirming the critical role of IL-1β in limiting S. aureus outgrowth during craniotomy infection. CONCLUSIONS These results demonstrate the existence of an initial anti-bacterial response that depends on both TLR2 and caspase-1 in controlling S. aureus growth; however, neither pathway is effective at clearing infection in the WT setting, since craniotomy infection persists when both molecules are present.
Collapse
Affiliation(s)
- Amy L Aldrich
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA
- Present Address: Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Shi
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rachel W Fallet
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
15
|
Aldrich A, Kuss MA, Duan B, Kielian T. 3D Bioprinted Scaffolds Containing Viable Macrophages and Antibiotics Promote Clearance of Staphylococcus aureus Craniotomy-Associated Biofilm Infection. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12298-12307. [PMID: 30855125 PMCID: PMC7034353 DOI: 10.1021/acsami.9b00264] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Craniotomy involves the removal of a skull fragment to access the brain, such as during tumor or epilepsy surgery, which is immediately replaced intraoperatively. The infection incidence after craniotomy ranges from 0.8 to 3%, with approximately half caused by Staphylococcus aureus ( S. aureus). To mitigate infectious complications following craniotomy, we engineered a three-dimensional (3D) bioprinted bone scaffold to harness the potent antibacterial activity of macrophages (MΦs) together with antibiotics using a mouse S. aureus craniotomy-associated biofilm model that establishes a persistent infection on the bone flap, subcutaneous galea, and brain. The 3D scaffold contained rifampin and daptomycin printed in a composite slurry, with viable MΦs incorporated into a hydrogel-based bioink, which was assessed for both the treatment and prevention of craniotomy-associated infections in the mouse model. For the treatment paradigm, the bone flap was removed at day 7 post infection after a mature biofilm had formed and was replaced with a 3D printed antibiotic scaffold, with or without MΦ incorporation. Bacterial burdens in the galea and brain were reduced by at least 100-fold at early time points, which was potentiated by bioprinting viable MΦs into the 3D antibiotic scaffold. We also examined a prevention paradigm, where the scaffolds were placed at the time of surgery and challenged with S. aureus one day later at the surgical site. Interestingly, unlike the treatment paradigm, the incorporation of viable MΦs into the 3D antibiotic scaffold did not enhance bacterial clearance compared to that of antibiotic alone. With further refinement, our 3D bioprinted scaffold represents a potential treatment modality, as it delivers therapeutic antibiotic levels more rapidly than systemic administration, based on its proximity to the infection site. In addition, the incorporation of viable MΦs into the 3D scaffold is an important advance, which demonstrated an improved therapeutic benefit for the treatment of established biofilms that represent the most clinically challenging scenario.
Collapse
Affiliation(s)
- Amy Aldrich
- Department of Pathology and Microbiology University of Nebraska Medical Center, Omaha, NE 68198
| | - Mitchell A. Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
16
|
Inflammatory and Immune-Mediated Cutaneous Diseases. Mediators Inflamm 2017; 2017:6793968. [PMID: 29445258 PMCID: PMC5763210 DOI: 10.1155/2017/6793968] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 11/18/2022] Open
|
17
|
Gries CM, Kielian T. Staphylococcal Biofilms and Immune Polarization During Prosthetic Joint Infection. J Am Acad Orthop Surg 2017; 25 Suppl 1:S20-S24. [PMID: 27922945 PMCID: PMC5640443 DOI: 10.5435/jaaos-d-16-00636] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Staphylococcal species are a leading cause of community- and nosocomial-acquired infections, where the placement of foreign materials increases infection risk. Indwelling medical devices and prosthetic implants are targets for staphylococcal cell adherence and biofilm formation. Biofilm products actively suppress proinflammatory microbicidal responses, as evident by macrophage polarization toward an anti-inflammatory phenotype and the recruitment of myeloid-derived suppressor cells. With the rise in prosthetic hip and knee arthroplasty procedures, together with the recalcitrance of biofilm infections to antibiotic therapy, it is imperative to better understand the mechanism of crosstalk between biofilm-associated bacteria and host immune cells. This review describes the current understanding of how staphylococcal biofilms evade immune-mediated clearance to establish persistent infections. The findings described herein may facilitate the identification of novel treatments for these devastating biofilm-mediated infections.
Collapse
Affiliation(s)
- Casey M. Gries
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
18
|
Scherr TD, Heim CE, Morrison JM, Kielian T. Hiding in Plain Sight: Interplay between Staphylococcal Biofilms and Host Immunity. Front Immunol 2014; 5:37. [PMID: 24550921 PMCID: PMC3913997 DOI: 10.3389/fimmu.2014.00037] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/22/2014] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus and Staphylococcus epidermidis are notable for their propensity to form biofilms on implanted medical devices. Staphylococcal biofilm infections are typified by their recalcitrance to antibiotics and ability to circumvent host immune-mediated clearance, resulting in the establishment of chronic infections that are often recurrent in nature. Indeed, the immunomodulatory lifestyle of biofilms seemingly shapes the host immune response to ensure biofilm engraftment and persistence in an immune competent host. Here, we provide a brief review of the mechanisms whereby S. aureus and S. epidermidis biofilms manipulate host–pathogen interactions and discuss the concept of microenvironment maintenance in infectious outcomes, as well as speculate how these findings pertain to the challenges of staphylococcal vaccine development.
Collapse
Affiliation(s)
- Tyler D Scherr
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - John M Morrison
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center , Omaha, NE , USA
| |
Collapse
|