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
|
Aili Y, Wei P, Yu X, Fan G, Maimaitiaili N, Li Y, Liu S, Huang Y, Zhao B, Wang Z, Qin H, Wang Y. Janus adhesive bio-patches with targeted drug delivery enabled anti-bacteria and pro-angiogenesis for dura mater repair. Mater Today Bio 2025; 31:101484. [PMID: 39925716 PMCID: PMC11804716 DOI: 10.1016/j.mtbio.2025.101484] [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: 10/23/2024] [Revised: 12/18/2024] [Accepted: 01/11/2025] [Indexed: 02/11/2025] Open
Abstract
Dural injuries often result in severe complications such as cerebrospinal fluid (CSF) leakage, intracranial infections, and brain herniation, which significantly impact patient recovery and quality of life. Conventional dural repair materials, which rely on suturing to peripheral tissues, fail to promote tissue regeneration and provide sufficient CSF leakage prevention, leading to suboptimal outcomes. To address these limitations, we developed a Janus adhesive bio-patch with both antibacterial and pro-angiogenic properties to enhance dura mater repair. This bio-patch consisted of a polyacrylic acid (PAA) adhesive gel layer loaded with vancomycin and magnesium carbonate (MgCO3), integrated onto a small intestinal submucosa (SIS) extracellular matrix. It exhibited a burst strength of 20.50±2.89kPa, effectively sealing CSF leaks, while demonstrating excellent antibacterial efficacy (∼99%) and significant enhanced angiogenesis (3.47-fold higher than the control). In rat, rabbit, and dog dural injury models, the bio-patch adhered seamlessly to the injury site, successfully preventing leaks and promoting tissue regeneration. These results highlighted the Janus adhesive bio-patch as a promising solution for improving dural repair in neurosurgery, offering a safer and more effective alternative to conventional suturing techniques.
Collapse
Affiliation(s)
- Yirizhati Aili
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System Tumors, Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| | - Pengfei Wei
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Xueqiao Yu
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Guofeng Fan
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| | - Nuerailijiang Maimaitiaili
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| | - Yunhuan Li
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Siqi Liu
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Yiqian Huang
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Bo Zhao
- Beijing Biosis Healing Biological Technology Co., Ltd, No.29 Yongda Road, Beijing, 102600, China
| | - Zengliang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System Tumors, Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| | - Hu Qin
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| | - Yongxin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
- Key Laboratory of Precision Diagnosis and Clinical Transformation of Nervous System Tumors, Xinjiang Medical University, No.393 Xinyi Road, Urumqi, Xinjiang, 830054, China
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
Van Roy Z, Kielian T. Tumor necrosis factor regulates leukocyte recruitment but not bacterial persistence during Staphylococcus aureus craniotomy infection. J Neuroinflammation 2024; 21:179. [PMID: 39044282 PMCID: PMC11264501 DOI: 10.1186/s12974-024-03174-9] [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: 03/12/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Craniotomy is a common neurosurgery used to treat intracranial pathologies. Nearly 5% of the 14 million craniotomies performed worldwide each year become infected, most often with Staphylococcus aureus (S. aureus), which forms a biofilm on the surface of the resected bone segment to establish a chronic infection that is recalcitrant to antibiotics and immune-mediated clearance. Tumor necrosis factor (TNF), a prototypical proinflammatory cytokine, has been implicated in generating protective immunity to various infections. Although TNF is elevated during S. aureus craniotomy infection, its functional importance in regulating disease pathogenesis has not been explored. METHODS A mouse model of S. aureus craniotomy infection was used to investigate the functional importance of TNF signaling using TNF, TNFR1, and TNFR2 knockout (KO) mice by quantifying bacterial burden, immune infiltrates, inflammatory mediators, and transcriptional changes by RNA-seq. Complementary experiments examined neutrophil extracellular trap formation, leukocyte apoptosis, phagocytosis, and bactericidal activity. RESULTS TNF transiently regulated neutrophil and granulocytic myeloid-derived suppressor cell recruitment to the brain, subcutaneous galea, and bone flap as evident by significant reductions in both cell types between days 7 to 14 post-infection coinciding with significant decreases in several chemokines, which recovered to wild type levels by day 28. Despite these defects, bacterial burdens were similar in TNF KO and WT mice. RNA-seq revealed enhanced lymphotoxin-α (Lta) expression in TNF KO granulocytes. Since both TNF and LTα signal through TNFR1 and TNFR2, KO mice for each receptor were examined to assess potential redundancy; however, neither strain had any impact on S. aureus burden. In vitro studies revealed that TNF loss selectively altered macrophage responses to S. aureus since TNF KO macrophages displayed significant reductions in phagocytosis, apoptosis, IL-6 production, and bactericidal activity in response to live S. aureus, whereas granulocytes were not affected. CONCLUSION These findings implicate TNF in modulating granulocyte recruitment during acute craniotomy infection via secondary effects on chemokine production and identify macrophages as a key cellular target of TNF action. However, the lack of changes in bacterial burden in TNF KO animals suggests the involvement of additional signals that dictate S. aureus pathogenesis during craniotomy infection.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
10
|
Gallucci S. DNA at the center of mammalian innate immune recognition of bacterial biofilms. Trends Immunol 2024; 45:103-112. [PMID: 38281884 PMCID: PMC11032746 DOI: 10.1016/j.it.2023.12.004] [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: 12/08/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/30/2024]
Abstract
Historically, the study of innate immune detection of bacterial infections has focused on the recognition of pathogen-associated molecular patterns (PAMPs) from bacteria growing as single cells in planktonic phase. However, over the past two decades, studies have highlighted an adaptive advantage of bacteria: the formation of biofilms. These structures are complex fortresses that stand against a hostile environment, including antibiotics and immune responses. Extracellular DNA (eDNA) is a crucial component of the matrix of most known biofilms. In this opinion article, I propose that eDNA is a universal PAMP that the immune system uses to recognize biofilms. Outstanding questions concern the discrimination between biofilm-associated eDNA and DNA from planktonic bacteria, the innate receptors involved, and the immune response to biofilms.
Collapse
Affiliation(s)
- Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Division of Innate Immunity, Department of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Zhang W, Kuss M, Yan Y, Shi W. Dynamic Alginate Hydrogel as an Antioxidative Bioink for Bioprinting. Gels 2023; 9:gels9040312. [PMID: 37102924 PMCID: PMC10137987 DOI: 10.3390/gels9040312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
3D bioprinting holds great potential for use in tissue engineering to treat degenerative joint disorders, such as osteoarthritis. However, there is a lack of multifunctional bioinks that can not only support cell growth and differentiation, but also offer protection to cells against injuries caused by the elevated oxidative stress; this conditions is a common characteristic of the microenvironment of the osteoarthritis disease. To mitigate oxidative stress-induced cellular phenotype change and malfunction, an anti-oxidative bioink derived from an alginate dynamic hydrogel was developed in this study. The alginate dynamic hydrogel gelated quickly via the dynamic covalent bond between the phenylboronic acid modified alginate (Alg-PBA) and poly (vinyl alcohol) (PVA). It presented good self-healing and shear-thinning abilities because of the dynamic feature. The dynamic hydrogel supported long-term growth of mouse fibroblasts after stabilization with a secondary ionic crosslinking between introduced calcium ions and the carboxylate group in the alginate backbone. In addition, the dynamic hydrogel showed good printability, resulting in the fabrication of scaffolds with cylindrical and grid structures with good structural fidelity. Encapsulated mouse chondrocytes maintained high viability for at least 7 days in the bioprinted hydrogel after ionic crosslinking. Most importantly, in vitro studies implied that the bioprinted scaffold could reduce the intracellular oxidative stress for embedded chondrocytes under H2O2 exposure; it could also protect the chondrocytes from H2O2-induced downregulation of extracellular matrix (ECM) relevant anabolic genes (ACAN and COL2) and upregulation of a catabolic gene (MMP13). In summary, the results suggest that the dynamic alginate hydrogel can be applied as a versatile bioink for the fabrication of 3D bioprinted scaffolds with an innate antioxidative ability; this technique is expected to improve the regenerative efficacy of cartilage tissues for the treatment of joint disorders.
Collapse
Affiliation(s)
- Wenhai Zhang
- Orthopedic Department, Tianjin Hospital, Tianjin 300211, China
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Yan
- Healthcare Security Office, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
14
|
Petronglo JR, Putnam NE, Ford CA, Cruz-Victorio V, Curry JM, Butrico CE, Fulbright LE, Johnson JR, Peck SH, Fatah SR, Cassat JE. Context-Dependent Roles for Toll-Like Receptors 2 and 9 in the Pathogenesis of Staphylococcus aureus Osteomyelitis. Infect Immun 2022; 90:e0041722. [PMID: 36226943 PMCID: PMC9670883 DOI: 10.1128/iai.00417-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is the major causative agent of bacterial osteomyelitis, an invasive infection of bone. Inflammation generated by the immune response to S. aureus contributes to bone damage by altering bone homeostasis. Increases in the differentiation of monocyte lineage cells into bone-resorbing osteoclasts (osteoclastogenesis) promote bone loss in the setting of osteomyelitis. In this study, we sought to define the role of Toll-like receptor (TLR) signaling in the pathogenesis of S. aureus osteomyelitis. We hypothesized that S. aureus-sensing TLRs 2 and 9, both of which are known to alter osteoclastogenesis in vitro, promote pathological changes to bone, including increased osteoclast abundance, bone loss, and altered callus formation during osteomyelitis. Stimulation of osteoclast precursors with S. aureus supernatant increased osteoclastogenesis in a TLR2-dependent, but not a TLR9-dependent, manner. However, in vivo studies using a posttraumatic murine model of osteomyelitis revealed that TLR2-null mice experienced similar bone damage and increased osteoclastogenesis compared to wild type (WT) mice. Therefore, we tested the hypothesis that compensation between TLR2 and TLR9 contributes to osteomyelitis pathogenesis. We found that mice deficient in both TLR2 and TLR9 (Tlr2/9-/-) have decreased trabecular bone loss in response to infection compared to WT mice. However, osteoclastogenesis is comparable between WT and Tlr2/9-/- mice, suggesting that alternative mechanisms enhance osteoclastogenesis in vivo during osteomyelitis. Indeed, we discovered that osteoclast precursors intracellularly infected with S. aureus undergo significantly increased osteoclast formation, even in the absence of TLR2 and TLR9. These results suggest that TLR2 and TLR9 have context-dependent roles in the alteration of bone homeostasis during osteomyelitis.
Collapse
Affiliation(s)
- Jenna R. Petronglo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Nicole E. Putnam
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Virginia Cruz-Victorio
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Jacob M. Curry
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Laura E. Fulbright
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Joshua R. Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Sun H. Peck
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - Sana R. Fatah
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Centergrid.412807.8, Nashville, Tennessee, USA
| |
Collapse
|
15
|
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
|
16
|
Daptomycin exerts differential immunomodulatory effects on host responses against methicillin-resistant Staphylococcus aureus biofilms. Int J Antimicrob Agents 2022; 60:106666. [PMID: 36038095 DOI: 10.1016/j.ijantimicag.2022.106666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Daptomycin (DAP) is indicated for difficult-to-treat Gram-positive infections, especially those caused by methicillin-resistant S. aureus (MRSA). Exposure of S. aureus to sub-inhibitory concentrations (sub-MICs) of antibiotics have been shown to alter cell morphology or biofilm formation. OBJECTIVES To investigate the influence of DAP biofilm sub-MICs on the damage caused by human polymorphonuclear neutrophils (PMNs) against MRSA biofilms and the potential immunomodulatory activity of DAP on human monocytes (MNCs) exposed to MRSA biofilms. METHODS DAP activity against biofilms and the impact of DAP on the PMNs-induced biofilm damage were evaluated by the XTT reduction assay, whereas pathogen recognition, signal transduction and cytokine modulation of DAP on MNCs in response to MRSA biofilms were assessed by RT-PCR and ELISA methodology. RESULTS The MIC50 of DAP to MRSA biofilms was 16 to 32 mg/L. Pre-treatment of MRSA to 1, 2 or 4 mg/L DAP caused a synergistic effect on PMN-mediated biofilm damage, being dependent on the effector-to-target ratio. MNCs responded to MRSA biofilms and DAP through Toll like receptor 2 (TLR2) upregulation and increased NLRP3 inflammasome production. DAP caused 2.5-fold greater TLR2 mRNA levels than those caused by MRSA biofilms. A predominantly inflammatory response was induced by either component, causing the release of significantly increased IFN-γ, TNF-α, IL-8 and IL-6 levels by MNCs exposed to the combination treatment. MRSA biofilms alone or combined with DAP caused low amounts of IL-10 production, but increased IL-1β levels. CONCLUSIONS DAP may condition MNCs towards an inflammatory response through TLR2 engagement and NLRP3 inflammasome activation, possibly controlling biofilm-associated pathogenicity.
Collapse
|
17
|
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: 2.0] [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
|
18
|
Dias ML, O'Connor KM, Dempsey EM, O'Halloran KD, McDonald FB. Targeting the Toll-like receptor pathway as a therapeutic strategy for neonatal infection. Am J Physiol Regul Integr Comp Physiol 2021; 321:R879-R902. [PMID: 34612068 DOI: 10.1152/ajpregu.00307.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Toll-like receptors (TLRs) are crucial transmembrane receptors that form part of the innate immune response. They play a role in the recognition of various microorganisms and their elimination from the host. TLRs have been proposed as vital immunomodulators in the regulation of multiple neonatal stressors that extend beyond infection such as oxidative stress and pain. The immune system is immature at birth and takes some time to become fully established. As such, babies are especially vulnerable to sepsis at this early stage of life. Findings suggest a gestational age-dependent increase in TLR expression. TLRs engage with accessory and adaptor proteins to facilitate recognition of pathogens and their activation of the receptor. TLRs are generally upregulated during infection and promote the transcription and release of proinflammatory cytokines. Several studies report that TLRs are epigenetically modulated by chromatin changes and promoter methylation upon bacterial infection that have long-term influences on immune responses. TLR activation is reported to modulate cardiorespiratory responses during infection and may play a key role in driving homeostatic instability observed during sepsis. Although complex, TLR signaling and downstream pathways are potential therapeutic targets in the treatment of neonatal diseases. By reviewing the expression and function of key Toll-like receptors, we aim to provide an important framework to understand the functional role of these receptors in response to stress and infection in premature infants.
Collapse
Affiliation(s)
- Maria L Dias
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland.,Department of Pediatrics and Child Health, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland
| | - Fiona B McDonald
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
TLR Signaling in Brain Immunity. Handb Exp Pharmacol 2021; 276:213-237. [PMID: 34761292 DOI: 10.1007/164_2021_542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Toll-like receptors (TLRs) comprise a group of transmembrane proteins with crucial roles in pathogen recognition, immune responses, and signal transduction. This family represented the first line of immune homeostasis in an evolutionarily conserved manner. Extensive researches in the past two decades had emphasized their structural and functional characteristics under both healthy and pathological conditions. In this review, we summarized the current understanding of TLR signaling in the central nervous system (CNS), which had been viewed as a previously "immune-privileged" but now "immune-specialized" area, with major implications for further investigation of pathological nature as well as potential therapeutic manipulation of TLR signaling in various neurological disorders.
Collapse
|
20
|
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
|
21
|
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
|
22
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
23
|
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
|