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
|
Liu ZH, Chen NY, Huang CY, Lin YJ, Yip PK, Wei KC, Liu HL. Modulation of the immune response by focused ultrasound suppressed brain abscess formation. Drug Deliv Transl Res 2025:10.1007/s13346-025-01847-3. [PMID: 40193008 DOI: 10.1007/s13346-025-01847-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2025] [Indexed: 04/12/2025]
Abstract
Brain abscess is a serious, life-threatening intracranial infection caused by inflammation and collection of infected material. Given the rise of multi-drug resistant strains and the widespread presence of bacteria, it is probable that the incidence of brain abscesses is expected to endure. The sequela of brain abscess constitutes a major source of morbidity and mortality. Brain abscess may cause permanent neurological damage, such as paresis, hydrocephalus, spasticity, mental deterioration and epileptic seizure. Current therapeutic approaches include surgical excision or drainage combined with prolonged antimicrobial treatment usually lasting 6-8 weeks. However, extended antimicrobial treatment may cause adverse side effects, such as nephrotoxicity, ototoxicity, and bone marrow suppression. As a result, it is essential to develop a novel approach to facilitate antibiotics delivery and shorten the therapeutic course clinically. Recently, focused ultrasound (FUS) has been demonstrated to have an ability to temporally open the brain blood barrier (BBB) and modulate the immune response in the brain tumor animal model or in naïve animals. In our study, we demonstrated the focused ultrasound treatment (3W acoustic power, 0.6 MPa peak negative pressure) to treat brain abscess by boosting immune response in CNS infection in the brain abscess animal model. The size of the brain abscess is reduced by 50 percent when the MRI scan is taken at 3 weeks post-treatment. The animals get better recovery after treatment. The use of low intensity FUS with systemic microbubble infusion to open the BBB by mechanical acoustic cavitation elicited an immediate immune response including elevations in proinflmmatory cytokine (IL-1, TNFα and IL-6) in the brain parenchyma surround the brain abscess. Furthermore, FUS exposure treatment also activated glial cells, potentially enhancing the encapsulation of brain abscesses and reducing the spread of bacteria to the adjacent brain parenchyma. Histological analysis also demonstrated that FUS can reduce neuron loss and blood vessel damage during brain abscess formation. Our findings indicate that the FUS system can achieve local reversible BBB opening, enhancing immunomodulation in an animal model of brain abscess.
Collapse
Affiliation(s)
- Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Nan-Yu Chen
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Chiung-Yin Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan
| | - Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan
| | - Ping K Yip
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County, 33305, Taiwan.
- Neuroscience Research Center, Chang Gung Memorial Hospital, Taoyuan, 33305, Taiwan.
| | - Hao-Li Liu
- Department of Electrical Engineering, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
3
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [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: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
4
|
Clegg J, Mnich ME, Carignano A, Cova G, Tavarini S, Sammicheli C, Clemente B, Smith M, Siena E, Bardelli M, Brazzoli M, Bagnoli F, McLoughlin RM, Soldaini E. Staphylococcus aureus-specific TIGIT + Treg are present in the blood of healthy subjects - a hurdle for vaccination? Front Immunol 2025; 15:1500696. [PMID: 39981298 PMCID: PMC11840346 DOI: 10.3389/fimmu.2024.1500696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/12/2024] [Indexed: 02/22/2025] Open
Abstract
Staphylococcus aureus poses an enormous burden of morbidity and mortality worldwide. Making an efficacious vaccine has however proven extremely challenging. Due to colonizing interactions, pre-existing S. aureus-specific CD4+ T cells are often found in the human population and yet a detailed characterization of their phenotypes and how they might in turn impact vaccine efficacy are thus far unknown. Using an activation induced marker assay to sort for S. aureus-specific CD4+ T cells in an effector function-independent manner, single cell transcriptomic analysis was conducted. Remarkably, S. aureus-specific CD4+ T cells consisted not only of a broader spectrum of conventional T cells (Tcon) than previously described but also of regulatory T cells (Treg). As compared to polyclonally-activated CD4+ T cells, S. aureus-specific Tcon were enriched for the expression of the Th17-type cytokine genes IL17A, IL22 and IL26, while higher percentages of S. aureus-specific Treg expressed the T Cell Immunoreceptor with Ig and ITIM domains (TIGIT), a pleiotropic immune checkpoint. Notably, the antagonistic anti-TIGIT mAb Tiragolumab increased IL-1β production in response to S. aureus in vitro. Therefore, these results uncover the presence of S. aureus-specific TIGIT+ Treg in the blood of healthy subjects that could blunt responses to vaccination and indicate TIGIT as a potential targetable biomarker to overcome pre-exposure-induced immunosuppression.
Collapse
Affiliation(s)
- Jonah Clegg
- GSK, Research Center, Siena, Italy
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Malgorzata E. Mnich
- GSK, Research Center, Siena, Italy
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | | | | | | | | | | | - Megan Smith
- GSK, Research Center, Siena, Italy
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | | - Rachel M. McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
5
|
Ghosh A, Gorain B. Mechanistic insight of neurodegeneration due to micro/nano-plastic-induced gut dysbiosis. Arch Toxicol 2025; 99:83-101. [PMID: 39370473 DOI: 10.1007/s00204-024-03875-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Despite offering significant conveniences, plastic materials contribute substantially in developing environmental hazards and pollutants. Plastic trash that has not been adequately managed may eventually break down into fragments caused by human or ecological factors. Arguably, the crucial element for determining the biological toxicities of plastics are micro/nano-forms of plastics (MPs/NPs), which infiltrate the mammalian tissue through different media and routes. Infiltration of MPs/NPs across the intestinal barrier leads to microbial architectural dysfunction, which further modulates the population of gastrointestinal microbes. Thereby, it triggers inflammatory mediators (e.g., IL-1α/β, TNF-α, and IFN-γ) by activating specific receptors located in the gut barrier. Mounting evidence indicates that MPs/NPs disrupt host pathophysiological function through modification of junctional proteins and effector cells. Moreover, the alteration of microbial diversity by MPs/NPs causes the breakdown of the blood-brain barrier and translocation of metabolites (e.g., SCFAs, LPS) through the vagus nerve. Potent penetration affects the neuronal networks, neuronal protein accumulation, acceleration of oxidative stress, and alteration of neurofibrillary tangles, and hinders distinctive communicating pathways. Conclusively, alterations of these neurotoxic factors are possibly responsible for the associated neurodegenerative disorders due to the exposure of MPs/NPs. In this review, the hypothesis on MPs/NPs associated with gut microbial dysbiosis has been interlinked to the distinct neurological impairment through the gut-brain axis.
Collapse
Affiliation(s)
- Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| |
Collapse
|
6
|
Mariager T, Bjarkam C, Nielsen H, Bodilsen J. Experimental animal models for brain abscess: a systematic review. Br J Neurosurg 2024; 38:1294-1301. [PMID: 36579498 DOI: 10.1080/02688697.2022.2160865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/04/2022] [Accepted: 10/18/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Brain abscess (BA) is a rare, but severe infection and experimental BA animal models may prove crucial for advances in treatment. This review describes the development of experimental BA models and the clinical advances obtained from these, in a historical perspective. MATERIAL AND METHODS Experimental BA studies from inception until June 15, 2022, were included by searching the PubMed and Embase databases. Inclusion required the use of an experimental BA animal model. Non-bacterial BA models, in vitro studies, veterinarian case-reports, and articles written in non-English language were excluded. Bias was not systematically assessed, and the review was not registered at the PROSPERO. RESULTS 79 studies were included. The majority of animal BA models have been based on small rodents using Staphylococcus aureus. The models have delineated the natural development of BA and provided detailed descriptions of the histopathological characteristics consisting of a necrotic centre surrounded by layers of inflammatory cells and fibroblasts encapsulated by a dense collagenous layer. Radiological studies of animal BA have been shown to correlate with the corresponding stages of human BA in both computed tomography and magnetic resonance imaging and may guide diagnosis as well as the timing of neurosurgical intervention. Moreover, pharmacokinetic studies of the intracavitary penetration of various antimicrobials have helped inform medical treatment of BA. Other studies have examined the diverse effects of corticosteroids including decreased cerebral oedema, intracranial pressure, and intracavitary drug concentration, whereas concerns on decreased or weakened capsule formation could not be confirmed. Finally, studies on the immunological response to BA have highlighted potential future immunomodulatory targets. CONCLUSIONS Animal models have been vital for improvements in the management of BA. Experimental BA models resembling human disease including polymicrobial infection by oral cavity flora in large animals are needed.
Collapse
Affiliation(s)
- Theis Mariager
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Carsten Bjarkam
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Nielsen
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
| | - Jacob Bodilsen
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
7
|
Han K, Wang X. Clinical efficacy and outcomes of calcitriol combined with bisphosphonates in the treatment of postmenopausal osteoporosis: A quasi-experimental study. Medicine (Baltimore) 2024; 103:e40171. [PMID: 39533562 PMCID: PMC11557120 DOI: 10.1097/md.0000000000040171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND A quasi-experimental study was conducted to investigate the clinical efficacy and outcomes of calcitriol combined with bisphosphonates in the treatment of postmenopausal osteoporosis (OP). METHODS A total of 152 patients with postmenopausal OP from March 2019 to June 2021 were enrolled. The patients who received calcitriol treatment were adopted as the control group, and the patients treated with calcitriol combined with bisphosphonates were considered as the intervention group. The treatment effects of patients were compared, and the pain degree of the joints of the patients was evaluated by Visual Analogue Scale/Score (VAS), the Barthel Index score was used to evaluate the daily living ability of patients, the hand and Oswestry Disability Index (ODI) were used to evaluate the dysfunction before and after treatment, and the bone metabolism indexes, immune cytokines and bone mineral density were detected before and after treatment, and the incidence of adverse reactions was calculated. RESULTS Regarding the therapeutic effects, the intervention group indicated an effective rate of 96.05% while the effective rate was 84.21% in the control group. The total effective rate of treatment in the intervention group was higher than the control group. The VAS, ODI scores, and bone metabolism indexes of the intervention group were significantly lower than the control group at 1, 2, and 3 months after treatment. The Barthel Index scores and bone mineral density of the intervention group were higher than the control group at 1, 2, and 3 months after treatment. The improvement of immune cytokines in the intervention group was significantly better than the control group (P < .05). One patient in the intervention group developed dizziness and 1 patient developed chills, with an adverse reaction rate of 2.63%, while in the control group, 2 patients had fever, and 2 patients developed chills, with an adverse reaction rate of 5.26% (P > .05). CONCLUSION Calcitriol combined with bisphosphonates has a significant clinical effect in the treatment of postmenopausal OP, which can significantly relieve bone pain in postmenopausal OP patients, enhance abnormal bone metabolism and immune function, and promote bone mineral density and daily living ability.
Collapse
Affiliation(s)
- Kui Han
- General Practice Department, Shanghai Changfeng Community Health Service Center of Putuo District, Shanghai, China
| | - Xiaoyan Wang
- General Practice Department, Shanghai Changfeng Community Health Service Center of Putuo District, Shanghai, China
| |
Collapse
|
8
|
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
|
9
|
Sakoulas G, Nizet V. Measuring beta-lactam minimum inhibitory concentrations in Staphylococcus aureus in the clinical microbiology laboratory: pinning the tail on the donkey. J Clin Microbiol 2024; 62:e0036623. [PMID: 37966224 PMCID: PMC10793257 DOI: 10.1128/jcm.00366-23] [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] [Indexed: 11/16/2023] Open
Abstract
Significant shortcomings have been identified in standard methods of susceptibility testing in bacteriological media, not only because the media fails to recapitulate the in vivo environment, but susceptibility testing itself fails to capture sub-MIC effects that significantly attenuate bacterial virulence properties. Until susceptibility testing conditions better recapitulate the in vivo environment, attempts to establish the quantitative relevance of beta-lactam MIC using current clinical microbiology standards in Staphylococcus aureus infections will likely prove unsuccessful.
Collapse
Affiliation(s)
- George Sakoulas
- Sharp Rees-Stealy Medical Group, San Diego, California, USA
- UCSD School of Medicine, La Jolla, California, USA
| | - Victor Nizet
- UCSD School of Medicine, La Jolla, California, USA
- Skaggs School of Pharmacy, UCSD School of Medicine, La Jolla, California, USA
| |
Collapse
|
10
|
Youn C, Pontaza C, Wang Y, Dikeman DA, Joyce DP, Alphonse MP, Wu MJ, Nolan SJ, Anany MA, Ahmadi M, Young J, Tocaj A, Garza LA, Wajant H, Miller LS, Archer NK. Neutrophil-intrinsic TNF receptor signaling orchestrates host defense against Staphylococcus aureus. SCIENCE ADVANCES 2023; 9:eadf8748. [PMID: 37327341 PMCID: PMC10275602 DOI: 10.1126/sciadv.adf8748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/11/2023] [Indexed: 06/18/2023]
Abstract
Staphylococcus aureus is the leading cause of skin and soft tissue infections and is a major health burden due to the emergence of antibiotic-resistant strains. To address the unmet need of alternative treatments to antibiotics, a better understanding of the protective immune mechanisms against S. aureus skin infection is warranted. Here, we report that tumor necrosis factor (TNF) promoted protection against S. aureus in the skin, which was mediated by bone marrow-derived immune cells. Furthermore, neutrophil-intrinsic TNF receptor (TNFR) signaling directed immunity against S. aureus skin infections. Mechanistically, TNFR1 promoted neutrophil recruitment to the skin, whereas TNFR2 prevented systemic bacterial dissemination and directed neutrophil antimicrobial functions. Treatment with a TNFR2 agonist showed therapeutic efficacy against S. aureus and Pseudomonas aeruginosa skin infections, which involved increased neutrophil extracellular trap formation. Our findings revealed nonredundant roles for TNFR1 and TNFR2 in neutrophils for immunity against S. aureus and can be therapeutically targeted for protection against bacterial skin infections.
Collapse
Affiliation(s)
- Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Cristina Pontaza
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Dustin A. Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Meng-Jen Wu
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Sabrina J. Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Mohamed A. Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg 97080, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, National Research Center, El Buhouth Street, Dokki, 12622 Giza, Egypt
| | - Michael Ahmadi
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Jeremy Young
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Aron Tocaj
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Luis A. Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg 97080, Germany
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| |
Collapse
|
11
|
Rani A, Toor D. Role of IL-1 in bacterial infections. MULTIFACETED ROLE OF IL-1 IN CANCER AND INFLAMMATION 2023:163-176. [DOI: 10.1016/b978-0-12-824273-5.00003-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Hassel B, Niehusmann P, Halvorsen B, Dahlberg D. Pro-inflammatory cytokines in cystic glioblastoma: A quantitative study with a comparison with bacterial brain abscesses. With an MRI investigation of displacement and destruction of the brain tissue surrounding a glioblastoma. Front Oncol 2022; 12:846674. [PMID: 35965529 PMCID: PMC9372434 DOI: 10.3389/fonc.2022.846674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic glioblastomas are aggressive primary brain tumors that may both destroy and displace the surrounding brain tissue as they grow. The mechanisms underlying these tumors’ destructive effect could include exposure of brain tissue to tumor-derived cytokines, but quantitative cytokine data are lacking. Here, we provide quantitative data on leukocyte markers and cytokines in the cyst fluid from 21 cystic glioblastomas, which we compare to values in 13 brain abscess pus samples. The concentration of macrophage/microglia markers sCD163 and MCP-1 was higher in glioblastoma cyst fluid than in brain abscess pus; lymphocyte marker sCD25 was similar in cyst fluid and pus, whereas neutrophil marker myeloperoxidase was higher in pus. Median cytokine levels in glioblastoma cyst fluid were high (pg/mL): TNF-α: 32, IL-6: 1064, IL-8: 23585, tissue factor: 28, the chemokine CXCL1: 639. These values were not significantly different from values in pus, pointing to a highly pro-inflammatory glioblastoma environment. In contrast, levels of IFN-γ, IL-1β, IL-2, IL-4, IL-10, IL-12, and IL-13 were higher in pus than in glioblastoma cyst fluid. Based on the quantitative data, we show for the first time that the concentrations of cytokines in glioblastoma cyst fluid correlate with blood leukocyte levels, suggesting an important interaction between glioblastomas and the circulation. Preoperative MRI of the cystic glioblastomas confirmed both destruction and displacement of brain tissue, but none of the cytokine levels correlated with degree of brain tissue displacement or peri-tumoral edema, as could be assessed by MRI. We conclude that cystic glioblastomas are highly pro-inflammatory environments that interact with the circulation and that they both displace and destroy brain tissue. These observations point to the need for neuroprotective strategies in glioblastoma therapy, which could include an anti-inflammatory approach.
Collapse
Affiliation(s)
- Bjørnar Hassel
- Department of Neurohabilitation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Defence Research Establishment (FFI), Kjeller, Norway
- *Correspondence: Bjørnar Hassel,
| | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Daniel Dahlberg
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Rodríguez AM, Rodríguez J, Giambartolomei GH. Microglia at the Crossroads of Pathogen-Induced Neuroinflammation. ASN Neuro 2022; 14:17590914221104566. [PMID: 35635133 PMCID: PMC9158411 DOI: 10.1177/17590914221104566] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Microglia are the resident tissue macrophages of the central nervous system (CNS). Recent findings point out that in the steady state the major role of microglia, is to instruct and regulate the correct function of the neuronal networks and different components of the neurovascular unit in the adult CNS, while providing immune surveillance. Paradoxically, during CNS infection immune activation of microglia generates an inflammatory milieu that contributes to the clearance of the pathogen but can, in the process, harm nearby cells of CNS. Most of the knowledge about the harmful effects of activated microglia on CNS has arisen from studies on neurodegenerative diseases. In this review we will focus on the beneficial role and detrimental functions of microglial cells on the neighboring cells of the CNS upon infection.
Collapse
Affiliation(s)
- Ana María Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julia Rodríguez
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Hernán Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. Facultad de Farmacia y Bioquímica, 28196Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Dhanagovind PT, Kujur PK, Swain RK, Banerjee S. IL-6 Signaling Protects Zebrafish Larvae during Staphylococcus epidermidis Infection in a Bath Immersion Model. THE JOURNAL OF IMMUNOLOGY 2021; 207:2129-2142. [PMID: 34544800 DOI: 10.4049/jimmunol.2000714] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/10/2021] [Indexed: 01/21/2023]
Abstract
The host immune responses to Staphylococcus epidermidis, a frequent cause of nosocomial infections, are not well understood. We have established a bath immersion model of this infection in zebrafish (Danio rerio) larvae. Macrophages play a primary role in the host immune response and are involved in clearance of infection in the larvae. S. epidermidis infection results in upregulation of tlr-2 There is marked inflammation characterized by heightened NF-κB signaling and elevation of several proinflammatory cytokines. There is rapid upregulation of il-1b and tnf-a transcripts, whereas an increase in il-6 levels is relatively more delayed. The IL-6 signaling pathway is further amplified by elevation of IL-6 signal transducer (il-6st) levels, which negatively correlates with miRNA dre-miR-142a-5p. Enhanced IL-6 signaling is protective to the host in this model as inhibition of the signaling pathway resulted in increased mortality upon S. epidermidis infection. Our study describes the host immune responses to S. epidermidis infection, establishes the importance of IL-6 signaling, and identifies a potential role of miR-142-5p-il-6st interaction in this infection model.
Collapse
Affiliation(s)
- P Thamarasseri Dhanagovind
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; and
| | - Prabeer K Kujur
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; and
| | | | - Sanjita Banerjee
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India; and
| |
Collapse
|
15
|
Alphonse MP, Rubens JH, Ortines RV, Orlando NA, Patel AM, Dikeman D, Wang Y, Vuong I, Joyce DP, Zhang J, Mumtaz M, Liu H, Liu Q, Youn C, Patrick GJ, Ravipati A, Miller RJ, Archer NK, Miller LS. Pan-caspase inhibition as a potential host-directed immunotherapy against MRSA and other bacterial skin infections. Sci Transl Med 2021; 13:13/601/eabe9887. [PMID: 34233954 DOI: 10.1126/scitranslmed.abe9887] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 03/02/2021] [Accepted: 05/26/2021] [Indexed: 01/01/2023]
Abstract
Staphylococcus aureus causes most skin infections in humans, and the emergence of methicillin-resistant S. aureus (MRSA) strains is a serious public health threat. There is an urgent clinical need for nonantibiotic immunotherapies to treat MRSA infections and prevent the spread of antibiotic resistance. Here, we investigated the pan-caspase inhibitor quinoline-valine-aspartic acid-difluorophenoxymethyl ketone (Q-VD-OPH) for efficacy against MRSA skin infection in mice. A single systemic dose of Q-VD-OPH decreased skin lesion sizes and reduced bacterial burden compared with vehicle-treated or untreated mice. Although Q-VD-OPH inhibited inflammasome-dependent apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC) speck formation and caspase-1-mediated interleukin-1β (IL-1β) production, Q-VD-OPH maintained efficacy in mice deficient in IL-1β, ASC, caspase-1, caspase-11, or gasdermin D. Thus, Q-VD-OPH efficacy was independent of inflammasome-mediated pyroptosis. Rather, Q-VD-OPH reduced apoptosis of monocytes and neutrophils. Moreover, Q-VD-OPH enhanced necroptosis of macrophages with concomitant increases in serum TNF and TNF-producing neutrophils, monocytes/macrophages, and neutrophils in the infected skin. Consistent with this, Q-VD-OPH lacked efficacy in mice deficient in TNF (with associated reduced neutrophil influx and necroptosis), in mice deficient in TNF/IL-1R and anti-TNF antibody-treated WT mice. In vitro studies revealed that combined caspase-3, caspase-8, and caspase-9 inhibition reduced apoptosis, and combined caspase-1, caspase-8, and caspase-11 inhibition increased TNF, suggesting a mechanism for Q-VD-OPH efficacy in vivo. Last, Q-VD-OPH also had a therapeutic effect against Streptococcus pyogenes and Pseudomonas aeruginosa skin infections in mice. Collectively, pan-caspase inhibition represents a potential host-directed immunotherapy against MRSA and other bacterial skin infections.
Collapse
Affiliation(s)
- Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jessica H Rubens
- Divison of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nicholas A Orlando
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Aman M Patel
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Dustin Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ivan Vuong
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mohammed Mumtaz
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Garrett J Patrick
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Advaitaa Ravipati
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
16
|
Liu H, Zhang S, Qiu M, Wang A, Ye J, Fu S. Garlic (Allium sativum) and Fu-ling (Poria cocos) mitigate lead toxicity by improving antioxidant defense mechanisms and chelating ability in the liver of grass carp (Ctenopharyngodon idella). ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:885-898. [PMID: 33830385 DOI: 10.1007/s10646-021-02405-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 06/12/2023]
Abstract
The heavy metal lead (Pb) is a contaminant widely distributed in the food chain. In this study, eight weeks of feeding containing Garlic (Allium sativum) or Fu-ling (Poria cocos) or both, markedly increased the growth index, enzyme activity, and serum index and significantly decreased muscle Pb level in grass carp (Ctenopharyngodon idella). Upon Pb exposure, the feeding Garlic or Fu-ling or both possessed the similar effects on improving the function of the antioxidant system and chelating ability. Further, the gene expressions of metal binding proteins (TF and MT-2) in the liver of the three experimental groups were significantly higher than those of the control group, which were all highly up-regulated after Pb exposure. At the same time, the activities of antioxidant enzymes (SOD and CAT) and the content of non-enzymatic substance (GSH) in the liver of the Garlic group, Fu-ling group and mixed group were stable compared to the control group after Pb exposure. Moreover, the reduction of Pb toxicity was manifested by the decrease of Pb content in the muscle, and the stable expression of heat stress proteins (HSP30 and HSP60) and immune-related genes (TNF-α and IL-1β). Taken together, the study preliminarily shows that the Garlic and Fu-ling play a role in mitigating the toxicity of Pb in grass carp.
Collapse
Affiliation(s)
- Haisu Liu
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Sanshan Zhang
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Ming Qiu
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Anli Wang
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Jianmin Ye
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Shengli Fu
- Guangdong Provincial Key Laboratory for Healthy and Saft Aquaculture, School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China.
| |
Collapse
|
17
|
Dey R, Bishayi B. Ciprofloxacin and dexamethasone in combination attenuate S. aureus induced brain abscess via neuroendocrine-immune interaction of TLR-2 and glucocorticoid receptor leading to behavioral improvement. Int Immunopharmacol 2021; 97:107695. [PMID: 33962227 DOI: 10.1016/j.intimp.2021.107695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus induced brain abscess is a critical health concern throughout the developing world. The conventional surgical intervention could not regulate the abscess-induced brain inflammation. Thus further study over the alternative therapeutic strategy for treating a brain abscess is of high priority. The resident glial cells recognize the invading S. aureus by their cell surface Toll-like receptor-2 (TLR-2). Glucocorticoid receptor (GR) was known for its immunosuppressive effects. In this study, an attempt had been taken to utilize the functional relationship or cross-talking between TLR-2 and GR during the pathogenesis of brain abscesses. Here, the combination of an antibiotic (i.e. ciprofloxacin) and dexamethasone was used to regulate the brain inflammation either in TLR-2 or GR blocking condition. We were also interested to figure out the possible impact of alternative therapy on behavioral impairments. The results indicated that combination treatment during TLR-2 blockade significantly reduced the bacterial burden and abscess area score in the infected brain. However, marked improvements were observed in anxiety, depression-like behavior, and motor co-ordination. The combination treatment after TLR-2 blocking effectively scavenged free radicals (H2O2, superoxide anion, and NO) through modulating antioxidant enzyme activities that ultimately control S. aureus induced glial reactivity possibly via up-regulating GR expression. The exogenous dexamethasone might regulate the GR expression in the brain by increasing the corticosterone concentration and the GC-GR mediated signaling. Therefore, this in-vivo study demonstrates the possible regulatory mechanism of bacterial brain abscess that involved TLR-2 and GR as a part of neuroendocrine-immune interaction.
Collapse
Affiliation(s)
- Rajen Dey
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India.
| |
Collapse
|
18
|
de Morais SD, Kak G, Menousek JP, Kielian T. Immunopathogenesis of Craniotomy Infection and Niche-Specific Immune Responses to Biofilm. Front Immunol 2021; 12:625467. [PMID: 33708216 PMCID: PMC7940520 DOI: 10.3389/fimmu.2021.625467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [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
|
19
|
The Relevance of IL-1-Signaling in the Protection against Gram-Positive Bacteria. Pathogens 2021; 10:pathogens10020132. [PMID: 33525468 PMCID: PMC7911888 DOI: 10.3390/pathogens10020132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/18/2021] [Accepted: 01/25/2021] [Indexed: 12/14/2022] Open
Abstract
Previous studies performed using a model of group B streptococcus (GBS)-induced peritoneal inflammation indicate that the interleukin-1 receptor (IL-1R) family plays an important role in the innate host defense against this encapsulated Gram-positive bacteria. Since the role of IL-1-dependent signaling in peritoneal infections induced by other Gram-positive bacteria is unknown, in the present study we sought to investigate the contribution of IL-1R signaling in host defenses against Streptococcus pyogenes (group A streptococcus or GAS) or Staphylococcus aureus, two frequent and global human Gram-positive extracellular pathogens. We analyzed here the outcome of GAS or S. aureus infection in IL-1R-deficient mice. After inoculated intraperitoneal (i.p.) inoculation with group A Streptococcus or S. aureus, all the wild-type (WT) control mice survived the challenge, while, respectively, 63% or 50% of IL-1-defective mice died. Lethality was due to the ability of both bacterial species to replicate and disseminate to the target organs of IL-1R-deficient mice. Moreover, the experimental results indicate that IL-1 signaling promotes the production of leukocyte attractant chemokines CXCL-1 and CXCL-2 and recruitment of neutrophils to bacterial infection sites. Accordingly, the reduced neutrophil recruitment in IL-1R-deficient mice was linked with decreased production of neutrophil chemokines. Collectively, our findings indicate that IL-1 signaling, as previously showed in host defense against GBS, plays a fundamental role also in controlling the progression and outcome of GAS or S. aureus disease.
Collapse
|
20
|
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: 171] [Impact Index Per Article: 42.8] [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
|
21
|
Wang Y, Ashbaugh AG, Dikeman DA, Zhang J, Ackerman NE, Kim SE, Falgons C, Ortines RV, Liu H, Joyce DP, Alphonse MP, Dillen CA, Thompson JM, Archer NK, Miller LS. Interleukin-1β and tumor necrosis factor are essential in controlling an experimental orthopedic implant-associated infection. J Orthop Res 2020; 38:1800-1809. [PMID: 31975434 PMCID: PMC7354231 DOI: 10.1002/jor.24608] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/15/2020] [Accepted: 01/22/2020] [Indexed: 02/04/2023]
Abstract
Orthopedic implant-associated infection (OIAI) is a major complication that leads to implant failure. In preclinical models of Staphylococcus aureus OIAI, osteomyelitis and septic arthritis, interleukin-1α (IL-1α), IL-1β, and tumor necrosis factor (TNF) are induced, but whether they have interactive or distinctive roles in host defense are unclear. Herein, a S. aureus OIAI model was performed in mice deficient in IL-1α, IL-1β, or TNF. Mice deficient in IL-1β or TNF (to a lesser extent) but not IL-1α had increased bacterial burden at the site of the OIAI throughout the 28-day experiment. IL-1β and TNF had a combined and critical role in host defense as mice deficient in both IL-1R and TNF (IL-1R/TNF-deficient mice) had a 40% mortality rate, which was associated with markedly increased bacterial burden at the site of the OIAI infection. Finally, IL-1α- and IL-1β-deficient mice had impaired neutrophil recruitment whereas IL-1β-, TNF-, and IL-1R/TNF-deficient mice all had impaired recruitment of both neutrophils and monocytes. Therefore, IL-1β and TNF contributed to host defense against S. aureus OIAI and neutrophil recruitment was primarily mediated by IL-1β and monocyte recruitment was mediated by both IL-1β and TNF.
Collapse
Affiliation(s)
- Yu Wang
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Alyssa G. Ashbaugh
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Dustin A. Dikeman
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Nicole E. Ackerman
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Sophie E. Kim
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Christian Falgons
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Roger V. Ortines
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Martin Prince Alphonse
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Carly A. Dillen
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - John M. Thompson
- Department of Orthopaedic Surgery, Johns Hopkins University
School of Medicine, Baltimore, MD, 21287, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School
of Medicine, Baltimore, MD, 21231, USA,Department of Orthopaedic Surgery, Johns Hopkins University
School of Medicine, Baltimore, MD, 21287, USA,Department of Medicine, Division of Infectious Diseases,
Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA,Department of Materials Science and Engineering, Johns
Hopkins University, Baltimore, MD, 21218, USA.,Janssen Research and Development, Spring House, PA, 19477,
USA.,Address correspondence to Lloyd S. Miller,
, Johns Hopkins Department of
Dermatology, Cancer Research Building II, Suite 205, 1550 Orleans Street,
Baltimore, MD 21231, Phone: (410) 955-8662, Fax: (410) 955-8645
| |
Collapse
|
22
|
Zhang Y, Hou B, Li C, Li H. Overexpression of circARF3 mitigates TNF-α-induced inflammatory damage by up-regulating miR-125b. Cell Cycle 2020; 19:1253-1264. [PMID: 32329660 DOI: 10.1080/15384101.2020.1731652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Encephalitis is the highest disability illness. We studied the function and mechanisms of circular RNA circARF3 (circARF3) in neurocyte cell inflammatory damage. CCK-8 assay and flow cytometry were, respectively, employed for examining the influences of tumor necrosis factor α (TNF-α), circARF3 and microRNA (miR)-125b on cell viability and apoptosis. The expression of circARF3 and miR-125b were changed by employing cell transfection and the results were determined by using qRT-PCR. Besides, the expression of Bcl-2, Bax, Cleaved-caspase-3, interleukin (IL)-1β, IL-6, IL-8 and cell pathways-related proteins were examined by using Western blot. The productions of IL-6, IL-8 and IL-1β were also tested by ELISA. The level of reactive oxygen species (ROS) was examined by ROS assay. We found that TNF-α caused inflammatory damage showing as suppressed cell viability, enhanced cell apoptosis, and increased cytokines production and ROS generation. Besides, TNF-α inducement also markedly reduced circARF3 expression. circARF3 overexpression mitigated TNF-α-induced cell inflammatory damage. Moreover, miR-125b was targeted and positively regulated by circARF3. Furthermore, miR-125b inhibition could reverse the influences of circARF3 overexpression. Besides, circARF3 restrained the JNK and NF-κB pathways by up-regulation of miR-125b. In conclusion, overexpression of circARF3 mitigated cell inflammatory damage via inactivation of JNK and NF-κB pathways and thereby up-regulation of miR-125b.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University , Qingdao, 266000, Shandong, China
| | - Binghui Hou
- Department of Neurology, The Affiliated Hospital of Qingdao University , Qingdao, 266000, Shandong, China
| | - Chunxiao Li
- Department of Neurology, The Affiliated Hospital of Qingdao University , Qingdao, 266000, Shandong, China
| | - Hong Li
- Department of Neurology, The Affiliated Hospital of Qingdao University , Qingdao, 266000, Shandong, China
| |
Collapse
|
23
|
Li B, Ding Y, Cheng X, Sheng D, Xu Z, Rong Q, Wu Y, Zhao H, Ji X, Zhang Y. Polyethylene microplastics affect the distribution of gut microbiota and inflammation development in mice. CHEMOSPHERE 2020; 244:125492. [PMID: 31809927 DOI: 10.1016/j.chemosphere.2019.125492] [Citation(s) in RCA: 345] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/28/2019] [Accepted: 11/26/2019] [Indexed: 05/23/2023]
Abstract
Environmental pollution caused by plastics has become a public health problem. However, the effect of microplastics on gut microbiota, inflammation development and their underlying mechanisms are not well characterized. In the present study, we assessed the effect of exposure to different amounts of polyethylene microplastics (6, 60, and 600 μg/day for 5 consecutive weeks) in a C57BL/6 mice model. Treatment with a high concentration of microplastics increased the numbers of gut microbial species, bacterial abundance, and flora diversity. Feeding groups showed a significant increase in Staphylococcus abundance alongside a significant decrease in Parabacteroides abundance, as compared to the blank (untreated) group. In addition, serum levels of interleukin-1α in all feeding groups were significantly greater than that in the blank group. Of note, treatment with microplastics decreased the percentage of Th17 and Treg cells among CD4+ cells, while no significant difference was observed between the blank and treatment groups with respect to the Th17/Treg cell ratio. The intestine (colon and duodenum) of mice fed high-concentration microplastics showed obvious inflammation and higher TLR4, AP-1, and IRF5 expression. Thus, polyethylene microplastics can induce intestinal dysbacteriosis and inflammation, which provides a theoretical basis for the prevention and treatment of microplastics-related diseases.
Collapse
Affiliation(s)
- Boqing Li
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yunfei Ding
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xue Cheng
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Dandan Sheng
- Yantai City Hospital for Infectious Diseases, No.62, Huanshan Road, Yantai, 264003, China
| | - Zheng Xu
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qianyu Rong
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yulong Wu
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Huilin Zhao
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiaofei Ji
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Ying Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
24
|
Wang X, Li X, Chen L, Yuan B, Liu T, Dong Q, Liu Y, Yin H. Interleukin-33 facilitates cutaneous defense against Staphylococcus aureus by promoting the development of neutrophil extracellular trap. Int Immunopharmacol 2020; 81:106256. [PMID: 32028244 DOI: 10.1016/j.intimp.2020.106256] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 12/23/2022]
Abstract
Neutrophil extracellular traps (NETs) are consisted of DNA fibers and granular proteins and the formation of NETs has been identified as a crucial element of innate immune defense. IL-33 is a member of the IL-1 family cytokines and has been known as a strong trigger of type-2 immunity. Growing studies imply that IL-33 is involved in host defense against microbial infection. Here, we investigate the underlying influence of IL-33 on NET formation in mice with S. aureus cutaneous infection. We found that the level of IL-33 was significantly elevated in skin lesions of S. aureus-infected mice. The alarmin IL-33 inspired host innate defense through activation of NADPH oxidase to produce reactive oxygen species (ROS). Besides mediating the direct bactericidal activity within phagolysosomes, ROS production in IL-33-primed neutrophils was also critical for induction of NET formation. Enhancement of NET production by IL-33 contributed to ensnaring S. aureus and bacterial killing activity in vitro and in vivo. All together, these findings set up an IL-33/ST2 axis modulating NET generation, which strengthens host defense of innate immunity against S. aureus infection.
Collapse
Affiliation(s)
- Xiaodi Wang
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiangyong Li
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Liying Chen
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Baohong Yuan
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qun Dong
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Hui Yin
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
25
|
Sakoulas G, Geriak M, Nizet V. Is a Reported Penicillin Allergy Sufficient Grounds to Forgo the Multidimensional Antimicrobial Benefits of β-Lactam Antibiotics? Clin Infect Dis 2020; 68:157-164. [PMID: 29986019 DOI: 10.1093/cid/ciy557] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 07/07/2018] [Indexed: 12/18/2022] Open
Abstract
The majority of patients with reported penicillin allergy are not allergic when tested or challenged. Penicillin allergy testing has been shown to significantly reduce annual healthcare expenditures. Data have emerged showing β-lactams have multidimensional antibacterial effects in vivo, far beyond what is appreciated in standard bacteriological susceptibility testing media. These include enhancing bacterial killing by the innate immune system. Supporting the clinical relevance of these secondary underappreciated effects are recent clinical and pharmacoeconomic analyses that show worse outcomes in patients with reported penicillin allergies who receive non-β-lactam antibiotics when compared to their non-penicillin-allergic counterparts. This is particularly relevant in the treatment of Staphylococcus aureus bacteremia. This article reviews the tremendous advantages offered by β-lactam therapy and makes a strong case that the debunking of false penicillin allergies through a detailed allergy history and penicillin allergy testing should be a vital component of antimicrobial stewardship practices.
Collapse
Affiliation(s)
- George Sakoulas
- Sharp Memorial Hospital, San Diego, La Jolla.,School of Medicine, University of California, San Diego, La Jolla
| | | | - Victor Nizet
- School of Medicine, University of California, San Diego, La Jolla.,Skaggs School of Pharmacy, University of California, San Diego, La Jolla
| |
Collapse
|
26
|
Liu C, Ouyang W, Xia J, Sun X, Zhao L, Xu F. Tumor Necrosis Factor-α Is Required for Mast Cell-Mediated Host Immunity Against Cutaneous Staphylococcus aureus Infection. J Infect Dis 2019; 218:64-74. [PMID: 29741644 DOI: 10.1093/infdis/jiy149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 04/17/2018] [Indexed: 01/09/2023] Open
Abstract
Background Mast cells (MCs) play a key role in immune process response to invading pathogens. Methods This study assessed the involvement of MCs in controlling Staphylococcus aureus infection in a cutaneous infection model of MC-deficient (KitW-sh/W-sh) mice. Results KitW-sh/W-sh mice developed significantly larger skin lesions after the cutaneous S. aureus challenge, when compared to wild-type (WT) mice, while MC dysfunction reduced the inflammation response to S. aureus. The levels of tumor necrosis factor (TNF)-α in skin tissues were significantly decreased in KitW-sh/W-sh mice upon infection. Moreover, the exogenous administration of MCs or recombinant TNF-α effectively restored the immune response against S. aureus in KitW-sh/W-sh mice via the recruitment of neutrophils to the infected site. These results indicate that the effects of MC deficiency are largely attributed to the decrease in production of TNF-α in cutaneous S. aureus infection. In addition, S. aureus-induced MC activation was dependent on the c-kit receptor-activated phosphoinositide 3-kinase (PI3K)/AKT/P65-nuclear factor (NF-κB) pathway, which was confirmed by treatment with Masitinib (a c-kit receptor inhibitor), Wortmannin (a PI3K inhibitor), and pyrrolidine dithiocarbamate (a NF-κB inhibitor), respectively. Conclusions The present study identifies the critical role of MCs in the host defense against S. aureus infection.
Collapse
Affiliation(s)
- Chao Liu
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Ouyang
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyan Xia
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoru Sun
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Liying Zhao
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Xu
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Dey R, Bishayi B. Dexamethasone exhibits its anti-inflammatory effects in S. aureus induced microglial inflammation via modulating TLR-2 and glucocorticoid receptor expression. Int Immunopharmacol 2019; 75:105806. [PMID: 31401378 DOI: 10.1016/j.intimp.2019.105806] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 01/31/2023]
Abstract
Microglial inflammation plays crucial role in the pathogenesis of CNS infections including brain abscesses. Staphylococcus aureus (S. aureus) is considered as one of the major causative agents of brain abscesses. Due to the emergence of multidrug resistant bacteria the available treatment options including conventional antibiotics and steroid therapy become ineffective in terms of inflammation regulation which warrants further investigation to resolve this health issue. Microglial TLR-2 plays important roles in the bacterial recognition as well as induction of inflammation whereas glucocorticoid receptor (GR) triggers anti-inflammatory pathways in presence of glucocorticoids (GCs). The main objective of this study was to figure out the interdependency between TLR-2 and GR in presence of exogenous dexamethasone during microglial inflammation as an alternative therapeutic approach. Experiments were done either in TLR-2 neutralized condition or GR blocked condition in presence of dexamethasone. Free radicals production, arginase, superoxide dismutase (SOD), catalase enzyme activities and corticosterone concentration were measured along with Western blot analysis of TLR-2, GR and other inflammatory molecules. The results suggested that dexamethasone pre-treatment in TLR-2 neutralized condition efficiently reduces the inflammatory consequences of S. aureus induced microglial inflammation through up regulating GR expression. During TLR-2 blocking dexamethasone exerted its potent anti-inflammatory activities via suppressing reactive oxygen species (ROS), NO production and up regulating arginase, SOD and catalase activities at the time point of 90 min. Further in-vivo experiments are needed to conclude that dexamethasone could resolve brain inflammation possibly through microglial phenotypic switching from pro-inflammatory M1 to anti-inflammatory M2.
Collapse
Affiliation(s)
- Rajen Dey
- Department of Physiology, Immunology Laboratory, University of Calcutta, University College of Science and Technology, Calcutta, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University College of Science and Technology, Calcutta, West Bengal, India.
| |
Collapse
|
28
|
Putnam NE, Fulbright LE, Curry JM, Ford CA, Petronglo JR, Hendrix AS, Cassat JE. MyD88 and IL-1R signaling drive antibacterial immunity and osteoclast-driven bone loss during Staphylococcus aureus osteomyelitis. PLoS Pathog 2019; 15:e1007744. [PMID: 30978245 PMCID: PMC6481883 DOI: 10.1371/journal.ppat.1007744] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/24/2019] [Accepted: 04/01/2019] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is able to infect virtually all organ systems and is a frequently isolated etiologic agent of osteomyelitis, a common and debilitating invasive infection of bone. Treatment of osteomyelitis requires invasive surgical procedures and prolonged antibiotic therapy, yet is frequently unsuccessful due to extensive pathogen-induced bone damage that can limit antibiotic penetration and immune cell influx to the infectious focus. We previously established that S. aureus triggers profound alterations in bone remodeling in a murine model of osteomyelitis, in part through the production of osteolytic toxins. However, staphylococcal strains lacking osteolytic toxins still incite significant bone destruction, suggesting that host immune responses are also major drivers of pathologic bone remodeling during osteomyelitis. The objective of this study was to identify host immune pathways that contribute to antibacterial immunity during S. aureus osteomyelitis, and to define how these immune responses alter bone homeostasis and contribute to bone destruction. We specifically focused on the interleukin-1 receptor (IL-1R) and downstream adapter protein MyD88 given the prominent role of this signaling pathway in both antibacterial immunity and osteo-immunologic crosstalk. We discovered that while IL-1R signaling is necessary for local control of bacterial replication during osteomyelitis, it also contributes to bone loss during infection. Mechanistically, we demonstrate that S. aureus enhances osteoclastogenesis of myeloid precursors in vitro, and increases the abundance of osteoclasts residing on bone surfaces in vivo. This enhanced osteoclast abundance translates to trabecular bone loss, and is dependent on intact IL-1R signaling. Collectively, these data define IL-1R signaling as a critical component of the host response to S. aureus osteomyelitis, but also demonstrate that IL-1R-dependent immune responses trigger collateral bone damage through activation of osteoclast-mediated bone resorption.
Collapse
Affiliation(s)
- Nicole E. Putnam
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Laura E. Fulbright
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jacob M. Curry
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Caleb A. Ford
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jenna R. Petronglo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Andrew S. Hendrix
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James E. Cassat
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
29
|
Abarca-Vargas R, Petricevich VL. Extract from Bougainvillea xbuttiana (Variety Orange) Inhibits Production of LPS-Induced Inflammatory Mediators in Macrophages and Exerts a Protective Effect In Vivo. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2034247. [PMID: 30949497 PMCID: PMC6425357 DOI: 10.1155/2019/2034247] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Different pharmacological properties, such as antioxidant, antiproliferative, and anti-inflammatory properties, have been described among natural products. We previously described that the Bougainvillea xbuttiana (Variety Orange) ethanolic extract (BxbO) has an anti-inflammatory effect; however, this action is not fully understood. In this study, the action of the BxbO extract on the secretion of inflammatory mediators in two experimental models, in vitro and in vivo, after LPS challenge was evaluated. METHODS Peritoneal macrophages were obtained from female BALB/c mice and LPS-challenged with or without the BxbO extract. For the evaluation of mediators, the supernatants at 0, 12, 24, 36, and 48 hours were collected. For in vivo estimation, groups of female BALB/c mice were first intraperitoneously injected with different amounts of LPS and later administered the oral BxbO extract (v.o.) for 144 hours. To understand the mechanism of action, sera obtained from mice were collected at 0, 2, 4, 8, 12, and 24 hours after LPS challenge (with or without BxbO) for the detection of mediators. RESULTS The results showed that, in both peritoneal macrophages and sera of mice treated with the BxbO extract 1 hour before or together with LPS challenge, proinflammatory cytokines and nitric oxide release were unquestionably repressed. In contrast, in both systems studied here, the IL-10 levels were elevated to 5 to 9 times. At lethal doses of LPS, the BxbO extract treatment was found to protect animals from death. CONCLUSIONS The results revealed that the inhibitory, protective, and benign effects of the BxbO extract were due to its capacity to balance the secretion of mediators.
Collapse
Affiliation(s)
- Rodolfo Abarca-Vargas
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Calle Leñeros Esquina Iztaccíhuatl s/n. Col. Volcanes, 62350 Cuernavaca, MOR, Mexico
| | - Vera L. Petricevich
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Calle Leñeros Esquina Iztaccíhuatl s/n. Col. Volcanes, 62350 Cuernavaca, MOR, Mexico
| |
Collapse
|
30
|
Lugo JM, Tafalla C, Oliva A, Pons T, Oliva B, Aquilino C, Morales R, Estrada MP. Evidence for antimicrobial and anticancer activity of pituitary adenylate cyclase-activating polypeptide (PACAP) from North African catfish (Clarias gariepinus): Its potential use as novel therapeutic agent in fish and humans. FISH & SHELLFISH IMMUNOLOGY 2019; 86:559-570. [PMID: 30481557 DOI: 10.1016/j.fsi.2018.11.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 06/09/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a regulatory neuropeptide that belongs to the secretin/glucagon superfamily, of which some members have shown antimicrobial activities. Contrasting to mammals, published studies on the action of PACAP in non-mammalian vertebrate immune system remain scarce. Some of our recent studies added this peptide to the growing list of mediators that allow cross-talk between the nervous, endocrine and immune systems in teleost fish. Regulation of PACAP and expression of its receptor genes has been demonstrated during an immune response mounted against acute bacterial infection in fish, though the direct effect of PACAP against fish pathogenic bacteria has never been addressed. Current work provides evidence of antimicrobial activity of Clarias gariepinus PACAP against a wide spectrum of Gram-negative and Gram-positive bacteria and fungi of interest for human medicine and aquaculture, in which computational prediction studies supported the putative PACAP therapeutic activity. Results also indicated that catfish PACAP not only exhibits inhibitory effects on pathogen growth, but also affects the proliferation of human non-small cell lung cancer cell line H460 in a dose-dependent manner. The observed cytotoxic activity of catfish PACAP against human tumor cells and pathogenic microorganisms, but not healthy fish and mammalian erythrocytes support a potential physiological role of this neuropeptide in selective microbial and cancer cell killing. All together, our findings extend the mechanisms by which PACAP could contribute to immune responses, and open up new avenues for future therapeutic application of this bioactive neuropeptide.
Collapse
Affiliation(s)
- Juana Maria Lugo
- Animal Biotechnology Department, Aquatic Biotechnology Group, Center for Genetic Engineering and Biotechnology, Havana, 10600, Cuba; Fish Immunology and Pathology Group, Animal Health Research Center (CISA-INIA), Valdeolmos, 28130, Madrid, Spain
| | - Carolina Tafalla
- Fish Immunology and Pathology Group, Animal Health Research Center (CISA-INIA), Valdeolmos, 28130, Madrid, Spain
| | - Ayme Oliva
- Animal Biotechnology Department, Veterinary Clinical Research Group, Center for Genetic Engineering and Biotechnology, Havana, 10600, Cuba
| | - Tirso Pons
- Structural Biology and BioComputing Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Brizaida Oliva
- Pharmaceutical Department. Laboratory of Cancer Biology. Center for Genetic Engineering and Biotechnology, Havana, 10600, Cuba
| | - Carolina Aquilino
- Fish Immunology and Pathology Group, Animal Health Research Center (CISA-INIA), Valdeolmos, 28130, Madrid, Spain
| | - Reynold Morales
- Animal Biotechnology Department, Aquatic Biotechnology Group, Center for Genetic Engineering and Biotechnology, Havana, 10600, Cuba
| | - Mario Pablo Estrada
- Animal Biotechnology Department, Aquatic Biotechnology Group, Center for Genetic Engineering and Biotechnology, Havana, 10600, Cuba.
| |
Collapse
|
31
|
An introduction to innate immunity in the central nervous system. ROLE OF INFLAMMATION IN ENVIRONMENTAL NEUROTOXICITY 2019. [DOI: 10.1016/bs.ant.2018.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Chen M, Low DCY, Low SYY, Muzumdar D, Seow WT. Management of brain abscesses: where are we now? Childs Nerv Syst 2018; 34:1871-1880. [PMID: 29968000 DOI: 10.1007/s00381-018-3886-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Brain abscesses affect all age groups and are not peculiar to a particular country, race, or geographical location. It is a disease that, in the past, carried a high morbidity and mortality. With improvements in medical technology and expertise, outcomes have improved tremendously. The causative organisms vary vastly and have evolved with time. Treatment of brain abscesses is primarily with antimicrobial therapy but surgery plays a vital role in achieving better outcomes. CONTENT In this article, we review the literature to find out how the epidemiology of this disease has changed through the years and re-visit the basic pathological process of abscess evolution and highlight the new research in the biochemical pathways that initiate and regulate this process. We also highlight how magnetic resonance imaging and its various modalities have improved diagnostic accuracy. Finally, we discuss the pros and cons of traditional open surgery versus newer minimally invasive methods.
Collapse
Affiliation(s)
- Minwei Chen
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore
| | - David C Y Low
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore.,Neurosurgical Service, KK Women's and Children's Hospital, Singapore, Singapore.,Singhealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore
| | - Sharon Y Y Low
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore.,Neurosurgical Service, KK Women's and Children's Hospital, Singapore, Singapore.,Singhealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore
| | - Dattatraya Muzumdar
- Department of Neurosurgery, King Edward VII Memorial hospital, Mumbai, India
| | - Wan Tew Seow
- Department of Neurosurgery, National Neuroscience Institute, Singapore, Singapore. .,Neurosurgical Service, KK Women's and Children's Hospital, Singapore, Singapore. .,Singhealth Duke-NUS Neuroscience Academic Clinical Program, Singapore, Singapore.
| |
Collapse
|
33
|
Wu X, Zhang Y, Chen X, Chen J, Jia M. Inflammatory immune response in rabbits with Staphylococcus aureus biofilm-associated sinusitis. Int Forum Allergy Rhinol 2018; 8:1226-1232. [PMID: 29979838 PMCID: PMC6282565 DOI: 10.1002/alr.22175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/21/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
Abstract
Background Staphylococcus aureus is the most commonly isolated bacterium from patients with surgically recalcitrant chronic rhinosinusitis (CRS). Understanding the immune responses to S aureus biofilms will provide insights into how the host response may be manipulated by therapeutic agents to improve the chances of successfully preventing and treating these infections. In this study, we investigated the inflammatory immune response in a rabbit model of S aureus biofilm–related sinusitis by analyzing the levels of some major inflammatory cytokines. Methods Eighteen New Zealand white rabbits were randomly divided into 3 groups: a blank‐control group; a negative‐control group; and a model group. Four weeks after the biofilm‐associated sinusitis models were established, the sinus mucosa was harvested and examined using hematoxylin‐eosin (H&E) staining, scanning electron microscopy (SEM), reverse transcription polymerase chain reaction (RT‐PCR), and western blotting. The expression levels of inflammatory cytokines were analyzed statistically. Results Interleukin (IL)‐1β, IL‐8, and tumor necrosis factor (TNF)‐α expression levels were significantly higher in the model group than in the blank‐control group (p < 0.05); mRNA levels were increased by 1600%, 230%, and 130%, respectively, and the protein levels were increased by 180%, 100%, and 100%, respectively. In contrast, IL‐4 and IL‐5 mRNA levels were reduced by 44% and 70%, respectively, compared with the blank‐control group (p < 0.05). Conclusion S aureus biofilms in the rabbit maxillary sinus mucosa were associated with increased IL‐1β, IL‐8, and TNF‐α expression, and decreased IL‐4 and IL‐5 expression.
Collapse
Affiliation(s)
- Xianmin Wu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Zhang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyun Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minghui Jia
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Gough P, Ganesan S, Datta SK. IL-20 Signaling in Activated Human Neutrophils Inhibits Neutrophil Migration and Function. THE JOURNAL OF IMMUNOLOGY 2017; 198:4373-4382. [PMID: 28424238 DOI: 10.4049/jimmunol.1700253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/23/2017] [Indexed: 12/23/2022]
Abstract
Neutrophils possess multiple antimicrobial mechanisms that are critical for protection of the host against infection with extracellular microbes, such as the bacterial pathogen Staphylococcus aureus Recruitment and activation of neutrophils at sites of infection are driven by cytokine and chemokine signals that directly target neutrophils via specific cell surface receptors. The IL-20 subfamily of cytokines has been reported to act at epithelial sites and contribute to psoriasis, wound healing, and anti-inflammatory effects during S. aureus infection. However, the ability of these cytokines to directly affect neutrophil function remains incompletely understood. In this article, we show that human neutrophils altered their expression of IL-20R chains upon migration and activation in vivo and in vitro. Such activation of neutrophils under conditions mimicking infection with S. aureus conferred responsiveness to IL-20 that manifested as modification of actin polymerization and inhibition of a broad range of actin-dependent functions, including phagocytosis, granule exocytosis, and migration. Consistent with the previously described homeostatic and anti-inflammatory properties of IL-20 on epithelial cells, the current study provides evidence that IL-20 directly targets and inhibits key inflammatory functions of neutrophils during infection with S. aureus.
Collapse
Affiliation(s)
- Portia Gough
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Sandip K Datta
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
35
|
Haroon E, Miller AH, Sanacora G. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology 2017; 42:193-215. [PMID: 27629368 PMCID: PMC5143501 DOI: 10.1038/npp.2016.199] [Citation(s) in RCA: 369] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/05/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
Increasing data indicate that inflammation and alterations in glutamate neurotransmission are two novel pathways to pathophysiology in mood disorders. The primary goal of this review is to illustrate how these two pathways may converge at the level of the glia to contribute to neuropsychiatric disease. We propose that a combination of failed clearance and exaggerated release of glutamate by glial cells during immune activation leads to glutamate increases and promotes aberrant extrasynaptic signaling through ionotropic and metabotropic glutamate receptors, ultimately resulting in synaptic dysfunction and loss. Furthermore, glutamate diffusion outside the synapse can lead to the loss of synaptic fidelity and specificity of neurotransmission, contributing to circuit dysfunction and behavioral pathology. This review examines the fundamental role of glia in the regulation of glutamate, followed by a description of the impact of inflammation on glial glutamate regulation at the cellular, molecular, and metabolic level. In addition, the role of these effects of inflammation on glia and glutamate in mood disorders will be discussed along with their translational implications.
Collapse
Affiliation(s)
- Ebrahim Haroon
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew H Miller
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
36
|
Hong X, Qin J, Li T, Dai Y, Wang Y, Liu Q, He L, Lu H, Gao Q, Lin Y, Li M. Staphylococcal Protein A Promotes Colonization and Immune Evasion of the Epidemic Healthcare-Associated MRSA ST239. Front Microbiol 2016; 7:951. [PMID: 27446000 PMCID: PMC4922140 DOI: 10.3389/fmicb.2016.00951] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/02/2016] [Indexed: 11/19/2022] Open
Abstract
The highly successful epidemic of healthcare-associated methicillin-resistant Staphylococcus aureus (HA-MRSA) ST239 is a growing concern worldwide, due to its progressive adaptation to the highly selective environment of the healthcare system. HA-MRSA ST239 display the reduced virulence and successfully colonize in hospital settings, while the emergent community-associated MRSA (CA-MRSA) maintain full virulence and cause infections in the community environment. Our aim was to investigate what enables S. aureus ST239 to be highly adaptive under hospital circumstances and gradually progress to a series of widespread invasive infections. We found that spa expression of HA-MRSA ST239 is much higher than that of CA-SA ST398. And we discovered that the highly production of staphylococcal protein A (SpA), having no concern with spa gene structure, enhances nasal colonization and cell adhesion in ST239. S. aureus ST239 defends against the adaptive immune response by resisting phagocytosis and inducing apoptosis of B cells through expression of surface-anchored and released protein A, facilitating its dissemination within the circulatory system to other organs. Protein A also plays another key role in subverting the host immune response through its ability to induce early shedding of TNF-α receptor 1 (TNFR1) from phagocytic cells. The increased levels of soluble TNFR1 present during experimental S. aureus ST239 infection may neutralize circulating TNF-α and impair the host inflammatory response. Protein A is also a virulence factor, as tested in our bacteremia model in mice, contributing to the durative tissue damage of abscess formation sites in ST239 infection. These functions of protein A eventually benefit to widespread infections of S. aureus ST239. We draw the conclusion that Staphylococcal Protein A may be a crucial determinant in the colonization and immune evasion of ST239 infections, contributing to persistent spread in the hospital settings. These results suggest that antibodies against protein A may provide insights into the development of novel treatments against S. aureus, especially HA-MRSA.
Collapse
Affiliation(s)
- Xufen Hong
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University Shanghai, China
| | - Juanxiu Qin
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Tianming Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yingxin Dai
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Qian Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Lei He
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Huiying Lu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Qianqian Gao
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yong Lin
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University Shanghai, China
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|
37
|
Contribution of Staphylococcus aureus Coagulases and Clumping Factor A to Abscess Formation in a Rabbit Model of Skin and Soft Tissue Infection. PLoS One 2016; 11:e0158293. [PMID: 27336691 PMCID: PMC4918888 DOI: 10.1371/journal.pone.0158293] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus produces numerous factors that facilitate survival in the human host. S. aureus coagulase (Coa) and von Willebrand factor-binding protein (vWbp) are known to clot plasma through activation of prothrombin and conversion of fibrinogen to fibrin. In addition, S. aureus clumping factor A (ClfA) binds fibrinogen and contributes to platelet aggregation via a fibrinogen- or complement-dependent mechanism. Here, we evaluated the contribution of Coa, vWbp and ClfA to S. aureus pathogenesis in a rabbit model of skin and soft tissue infection. Compared to skin abscesses caused by the Newman wild-type strain, those caused by isogenic coa, vwb, or clfA deletion strains, or a strain deficient in coa and vwb, were significantly smaller following subcutaneous inoculation in rabbits. Unexpectedly, we found that fibrin deposition and abscess capsule formation appear to be independent of S. aureus coagulase activity in the rabbit infection model. Similarities notwithstanding, S. aureus strains deficient in coa and vwb elicited reduced levels of several proinflammatory molecules in human blood in vitro. Although a specific mechanism remains to be determined, we conclude that S. aureus Coa, vWbp and ClfA contribute to abscess formation in rabbits.
Collapse
|
38
|
Greaney AJ, Leppla SH, Moayeri M. Bacterial Exotoxins and the Inflammasome. Front Immunol 2015; 6:570. [PMID: 26617605 PMCID: PMC4639612 DOI: 10.3389/fimmu.2015.00570] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/26/2015] [Indexed: 11/24/2022] Open
Abstract
The inflammasomes are intracellular protein complexes that play an important role in innate immune sensing. Activation of inflammasomes leads to activation of caspase-1 and maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. In certain myeloid cells, this activation can also lead to an inflammatory cell death (pyroptosis). Inflammasome sensor proteins have evolved to detect a range of microbial ligands and bacterial exotoxins either through direct interaction or by detection of host cell changes elicited by these effectors. Bacterial exotoxins activate the inflammasomes through diverse processes, including direct sensor cleavage, modulation of ion fluxes through plasma membrane pore formation, and perturbation of various host cell functions. In this review, we summarize the findings on some of the bacterial exotoxins that activate the inflammasomes.
Collapse
Affiliation(s)
- Allison J Greaney
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | - Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
39
|
Tumor necrosis factor-α and interleukin-1β gene polymorphisms and risk of brain abscess in North Indian population. Cytokine 2015; 75:159-64. [DOI: 10.1016/j.cyto.2015.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 07/07/2015] [Accepted: 07/08/2015] [Indexed: 12/30/2022]
|
40
|
Mehta D, Petes C, Gee K, Basta S. The Role of Virus Infection in Deregulating the Cytokine Response to Secondary Bacterial Infection. J Interferon Cytokine Res 2015; 35:925-34. [PMID: 26308503 DOI: 10.1089/jir.2015.0072] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Proinflammatory cytokines are produced by macrophages and dendritic cells (DCs) after infection to stimulate T helper (Th) cells, linking innate and adaptive immunity. Virus infections can deregulate the proinflammatory cytokine response like tumor necrosis factor-α and interleukin (IL)-2, making the host more susceptible to secondary bacterial infections. Studies using various viruses such as lymphocytic choriomeningitis virus, influenza A virus, and human immunodeficiency virus have revealed several intriguing mechanisms that account for the increased susceptibility to several prevalent bacterial infections. In particular, type I interferons induced during a virus infection have been observed to play a role in suppressing the production of some key antibacterial proinflammatory cytokines such as IL-23 and IL-17. Other suppressive mechanisms as a result of cytokine deregulation by viral infections include reduced function of immune cells such as DC, macrophage, natural killer, CD4(+), and CD8(+) T cells leading to impaired clearance of secondary bacterial infections. In this study, we highlight some of the immune mechanisms that become deregulated by viral infections, and can thus become defective during secondary bacterial infections.
Collapse
Affiliation(s)
- Divya Mehta
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Carlene Petes
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| |
Collapse
|
41
|
Mast Cell Serotonin Immunoregulatory Effects Impacting on Neuronal Function: Implications for Neurodegenerative and Psychiatric Disorders. Neurotox Res 2015; 28:147-53. [PMID: 26038194 DOI: 10.1007/s12640-015-9533-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/15/2015] [Accepted: 05/15/2015] [Indexed: 01/05/2023]
Abstract
Mast cells (MCs) are derived from hemopoietic precursor cells, undergo their maturation in peripheral tissues, and play a significant role in both the innate and adaptive immune response. Cross-linking of the FcεRI on MCs initiates activation of several cytoplasmic protein tyrosine kinases which rapidly lead to phosphorylation and recruitment of adaptor molecules. These effects trigger the release of preformed mediators stored in the cytoplasmic granules, including histamine, serotonin and tryptase, as well as newly synthesized mediators, such as cytokines/chemokines, prostaglandins, leukotrienes, and growth factors. Serotonin (5-HT) is a bioactive monoamine, which has seven specific cell surface membrane bound receptors which are coupled to G-proteins, plays an important role in the central and peripheral nervous system, and is one of the key mediators in signaling between nervous and immune systems. Serotonin is not stored in all MC types but is implicated in MC adhesion, chemotaxis, tumorigenesis, and tissue regeneration through smooth muscle differentiation of stromal cells. Recent evidence indicates that serotonin has immunoregulatory actions that may be important in neuropsychiatric conditions. Chemokines, RANTES/CCL5, MCP-1/CCL2, and related molecules, constitute the C-C class of chemokine supergene family, play a role in regulating T helper-cell cytokine production and MC trafficking, and are involved in histamine and serotonin generation and MC functions. Pro-inflammatory cytokines such as interleukin-1-β and tumor necrosis factor which mediate MC response, are capable of activating p38 MAPK, and might increase serotonin generation through p38 MAPK activation. Here, we review the relationship between MCs and serotonin and its role in inflammatory diseases and neuroimmune interactions.
Collapse
|
42
|
Kobayashi SD, Malachowa N, DeLeo FR. Pathogenesis of Staphylococcus aureus abscesses. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1518-27. [PMID: 25749135 DOI: 10.1016/j.ajpath.2014.11.030] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 11/13/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023]
Abstract
Staphylococcus aureus causes many types of human infections and syndromes-most notably skin and soft tissue infections. Abscesses are a frequent manifestation of S. aureus skin and soft tissue infections and are formed, in part, to contain the nidus of infection. Polymorphonuclear leukocytes (neutrophils) are the primary cellular host defense against S. aureus infections and a major component of S. aureus abscesses. These host cells contain and produce many antimicrobial agents that are effective at killing bacteria, but can also cause non-specific damage to host tissues and contribute to the formation of abscesses. By comparison, S. aureus produces several molecules that also contribute to the formation of abscesses. Such molecules include those that recruit neutrophils, cause host cell lysis, and are involved in the formation of the fibrin capsule surrounding the abscess. Herein, we review our current knowledge of the mechanisms and processes underlying the formation of S. aureus abscesses, including the involvement of polymorphonuclear leukocytes, and provide a brief overview of therapeutic approaches.
Collapse
Affiliation(s)
- Scott D Kobayashi
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Natalia Malachowa
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Frank R DeLeo
- Laboratory of Human Bacterial Pathogenesis, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana.
| |
Collapse
|
43
|
Feuerstein R, Seidl M, Prinz M, Henneke P. MyD88 in macrophages is critical for abscess resolution in staphylococcal skin infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2735-45. [PMID: 25681348 DOI: 10.4049/jimmunol.1402566] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
When Staphylococcus aureus penetrates the epidermis and reaches the dermis, polymorphonuclear leukocytes (PMLs) accumulate and an abscess is formed. However, the molecular mechanisms that orchestrate initiation and termination of inflammation in skin infection are incompletely understood. In human myeloid differentiation primary response gene 88 (MyD88) deficiency, staphylococcal skin and soft tissue infections are a leading and potentially life-threatening problem. In this study, we found that MyD88-dependent sensing of S. aureus by dermal macrophages (Mϕ) contributes to both timely escalation and termination of PML-mediated inflammation in a mouse model of staphylococcal skin infection. Mϕs were key to recruit PML within hours in response to staphylococci, irrespective of bacterial viability. In contrast with bone marrow-derived Mϕs, dermal Mϕs did not require UNC-93B or TLR2 for activation. Moreover, PMLs, once recruited, were highly activated in an MyD88-independent fashion, yet failed to clear the infection if Mϕs were missing or functionally impaired. In normal mice, clearance of the infection and contraction of the PML infiltrate were accompanied by expansion of resident Mϕs in a CCR2-dependent fashion. Thus, whereas monocytes were dispensable for the early immune response to staphylococci, they contributed to Mϕ renewal after the infection was overcome. Taken together, MyD88-dependent sensing of staphylococci by resident dermal Mϕs is key for a rapid and balanced immune response, and PMLs are dependent on intact Mϕ for full function. Renewal of resident Mϕs requires both local control of bacteria and inflammatory monocytes entering the skin.
Collapse
Affiliation(s)
- Reinhild Feuerstein
- Center for Chronic Immunodeficiency, University Medical Center, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Maximilian Seidl
- Center for Chronic Immunodeficiency, University Medical Center, University of Freiburg, 79106 Freiburg, Germany; Institute of Pathology, University Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center, University of Freiburg, 79106 Freiburg, Germany; Centre of Biological Signalling Studies, University of Freiburg, 79106 Freiburg, Germany; and
| | - Philipp Henneke
- Center for Chronic Immunodeficiency, University Medical Center, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, University Medical Center, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
44
|
Alsina L, Israelsson E, Altman MC, Dang KK, Ghandil P, Israel L, von Bernuth H, Baldwin N, Qin H, Jin Z, Banchereau R, Anguiano E, Ionan A, Abel L, Puel A, Picard C, Pascual V, Casanova JL, Chaussabel D. A narrow repertoire of transcriptional modules responsive to pyogenic bacteria is impaired in patients carrying loss-of-function mutations in MYD88 or IRAK4. Nat Immunol 2014; 15:1134-42. [PMID: 25344726 PMCID: PMC4281021 DOI: 10.1038/ni.3028] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/08/2014] [Indexed: 12/14/2022]
Abstract
Loss of function of the kinase IRAK4 or the adaptor MyD88 in humans interrupts a pathway critical for pathogen sensing and ignition of inflammation. However, patients with loss-of-function mutations in the genes encoding these factors are, unexpectedly, susceptible to only a limited range of pathogens. We employed a systems approach to investigate transcriptome responses following in vitro exposure of patients' blood to agonists of Toll-like receptors (TLRs) and receptors for interleukin 1 (IL-1Rs) and to whole pathogens. Responses to purified agonists were globally abolished, but variable residual responses were present following exposure to whole pathogens. Further delineation of the latter responses identified a narrow repertoire of transcriptional programs affected by loss of MyD88 function or IRAK4 function. Our work introduces the use of a systems approach for the global assessment of innate immune responses and the characterization of human primary immunodeficiencies.
Collapse
Affiliation(s)
- L Alsina
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
- Allergy and Clinical Immunology Department, Hospital Sant Joan de
Déu, Barcelona Universitat de Barcelona, Spain, EU
| | - E Israelsson
- Benaroya Research Institute at Virginia Mason, Seattle, Washington,
USA
| | - MC Altman
- Benaroya Research Institute at Virginia Mason, Seattle, Washington,
USA
| | - KK Dang
- Benaroya Research Institute at Virginia Mason, Seattle, Washington,
USA
| | - P Ghandil
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
| | - L Israel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
| | - H von Bernuth
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
- Department of Pediatric Pneumology and Immunology, Charité
Hospital– Humboldt University, Berlin, Germany, EU
| | - N Baldwin
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - H Qin
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - Z Jin
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - R Banchereau
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - E Anguiano
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - A Ionan
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - L Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
| | - A Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases,
Rockefeller Branch, The Rockefeller University, New York, USA
| | - C Picard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
- Study Center of Primary Immunodeficiencies, Assistance
Publique-Hôpitaux de Paris, Necker Hospital, Paris, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases,
Rockefeller Branch, The Rockefeller University, New York, USA
- Howard Hughes Medical Institute, New York, USA
| | - V Pascual
- Baylor Institute for Immunology Research and Baylor Research
Institute, Dallas, Texas, USA
| | - JL Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch,
INSERM UMR 1163, IMAGINE Institute, Paris, France, EU
- Paris Descartes University, France, EU
- St. Giles Laboratory of Human Genetics of Infectious Diseases,
Rockefeller Branch, The Rockefeller University, New York, USA
- Howard Hughes Medical Institute, New York, USA
- Pediatric Hematology-Immunology Unit, Necker Hospital, Assistance
Publique-Hôpitaux de Paris, Paris, France, EU
| | - D Chaussabel
- Benaroya Research Institute at Virginia Mason, Seattle, Washington,
USA
- Sidra Medical and Research Center, Doha, Qatar
| |
Collapse
|
45
|
The nuclear factor kappa B (NF-κB) activation is required for phagocytosis of staphylococcus aureus by RAW 264.7 cells. Exp Cell Res 2014; 327:256-63. [DOI: 10.1016/j.yexcr.2014.04.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 11/23/2022]
|
46
|
The interleukin-1β/CXCL1/2/neutrophil axis mediates host protection against group B streptococcal infection. Infect Immun 2014; 82:4508-17. [PMID: 25114117 DOI: 10.1128/iai.02104-14] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have indicated that group B streptococcus (GBS), a frequent human pathogen, potently induces the release of interleukin-1β (IL-1β), an important mediator of inflammatory responses. Since little is known about the role of this cytokine in GBS disease, we analyzed the outcome of infection in IL-1β-deficient mice. These animals were markedly sensitive to GBS infection, with most of them dying under challenge conditions that caused no deaths in wild-type control mice. Lethality was due to the inability of the IL-1β-deficient mice to control local GBS replication and dissemination to target organs, such as the brain and the kidneys. Moreover, in a model of inflammation induced by the intraperitoneal injection of killed GBS, a lack of IL-1β was associated with selective impairment in the production of the neutrophil chemokines CXCL1 and CXCL2 and in neutrophil recruitment to the peritoneal cavity. Decreased blood neutrophil counts and impaired neutrophil recruitment to the brain and kidneys were also observed during GBS infection in IL-1β-deficient mice concomitantly with a reduction in CXCL1 and CXCL2 tissue levels. Notably, the hypersusceptibility to GBS infection observed in the immune-deficient animals was recapitulated by neutrophil depletion with anti-Gr1 antibodies. Collectively, our data identify a cytokine circuit that involves IL-1β-induced production of CXCL1 and CXCL2 and leads the recruitment of neutrophils to GBS infection sites. Moreover, our data point to an essential role of these cells in controlling the progression and outcome of GBS disease.
Collapse
|
47
|
Hanamsagar R, Aldrich A, Kielian T. Critical role for the AIM2 inflammasome during acute CNS bacterial infection. J Neurochem 2014; 129:704-11. [PMID: 24484406 DOI: 10.1111/jnc.12669] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/26/2013] [Accepted: 01/27/2014] [Indexed: 01/16/2023]
Abstract
Interleukin-1β (IL-1β) is essential for eliciting protective immunity during the acute phase of Staphylococcus aureus (S. aureus) infection in the central nervous system (CNS). We previously demonstrated that microglial IL-1β production in response to live S. aureus is mediated through the Nod-like receptor protein 3 (NLRP3) inflammasome, including the adapter protein ASC (apoptosis-associated speck-like protein containing a caspase-1 recruitment domain), and pro-caspase 1. Here, we utilized NLRP3, ASC, and caspase 1/11 knockout (KO) mice to demonstrate the functional significance of inflammasome activity during CNS S. aureus infection. ASC and caspase 1/11 KO animals were exquisitely sensitive, with approximately 50% of mice succumbing to infection within 24 h. Unexpectedly, the survival of NLRP3 KO mice was similar to wild-type animals, suggesting the involvement of an alternative upstream sensor, which was later identified as absent in melanoma 2 (AIM2) based on the similar disease patterns between AIM2 and ASC KO mice. Besides IL-1β, other key inflammatory mediators, including IL-6, CXCL1, CXCL10, and CCL2 were significantly reduced in the CNS of AIM2 and ASC KO mice, implicating autocrine/paracrine actions of IL-1β, as these mediators do not require inflammasome processing for secretion. These studies demonstrate a novel role for the AIM2 inflammasome as a critical molecular platform for regulating IL-1β release and survival during acute CNS S. aureus infection.
Collapse
Affiliation(s)
- Richa Hanamsagar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | |
Collapse
|
48
|
Drew PD, Kane CJM. Fetal alcohol spectrum disorders and neuroimmune changes. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 118:41-80. [PMID: 25175861 DOI: 10.1016/b978-0-12-801284-0.00003-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The behavioral consequences of fetal alcohol spectrum disorders (FASD) are serious and persist throughout life. The causative mechanisms underlying FASD are poorly understood. However, much has been learned about FASD from human structural and functional studies as well as from animal models, which have provided a greater understanding of the mechanisms underlying FASD. Using animal models of FASD, it has been recently discovered that ethanol induces neuroimmune activation in the developing brain. The resulting microglial activation, production of proinflammatory molecules, and alteration in expression of developmental genes are postulated to alter neuron survival and function and lead to long-term neuropathological and cognitive defects. It has also been discovered that microglial loss occurs, reducing microglia's ability to protect neurons and contribute to neuronal development. This is important, because emerging evidence demonstrates that microglial depletion during brain development leads to long-term neuropathological and cognitive defects. Interestingly, the behavioral consequences of microglial depletion and neuroimmune activation in the fetal brain are particularly relevant to FASD. This chapter reviews the neuropathological and behavioral abnormalities of FASD and delineates correlates in animal models. This serves as a foundation to discuss the role of the neuroimmune system in normal brain development, the consequences of microglial depletion and neuroinflammation, the evidence of ethanol induction of neuroinflammatory processes in animal models of FASD, and the development of anti-inflammatory therapies as a new strategy for prevention or treatment of FASD. Together, this knowledge provides a framework for discussion and further investigation of the role of neuroimmune processes in FASD.
Collapse
Affiliation(s)
- Paul D Drew
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Cynthia J M Kane
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| |
Collapse
|
49
|
Yin H, Li X, Hu S, Liu T, Yuan B, Ni Q, Lan F, Luo X, Gu H, Zheng F. IL-33 promotes Staphylococcus aureus-infected wound healing in mice. Int Immunopharmacol 2013; 17:432-8. [DOI: 10.1016/j.intimp.2013.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/28/2013] [Accepted: 07/12/2013] [Indexed: 12/11/2022]
|
50
|
Nygaard TK, Pallister KB, Zurek OW, Voyich JM. The impact of α-toxin on host cell plasma membrane permeability and cytokine expression during human blood infection by CA-MRSA USA300. J Leukoc Biol 2013; 94:971-9. [PMID: 24026286 DOI: 10.1189/jlb.0213080] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This investigation examines the influence of α-toxin (Hla) expression by CA-MRSA on host immune cell integrity and cytokine expression during infection of human blood. Flow cytometry analysis of human blood infected by Staphylococcus aureus PFGE type USA300 or a USA300Δhla demonstrated that Hla expression significantly increased plasma membrane permeability of human CD14(+) monocytes. The increased susceptibility of human CD14(+) monocytes to Hla toxicity paralleled the high cell-surface expression on these cell types of ADAM10. USA300 rapidly associated with PMNs and monocytes but not T cells following inoculation of human blood. Transcription analysis indicated a strong up-regulation of proinflammatory cytokine transcription following infection of human blood by USA300 and USA300Δhla. CBAs and ELISAs determined that IL-6, IL-10, TNF-α, IFN-γ, IL-1β, IL-8, and IL-4 are significantly up-regulated during the initial phases of human blood infection by USA300 relative to mock-infected blood but failed to distinguish any significant differences in secreted cytokine protein concentrations during infection by USA300Δhla relative to USA300. Collectively, these findings demonstrate that expression of Hla by USA300 has a significant impact on human CD14(+) monocyte plasma membrane integrity but is not exclusively responsible for the proinflammatory cytokine profile induced by USA300 during the initial stages of human blood infection.
Collapse
Affiliation(s)
- Tyler K Nygaard
- 1.Lewis Hall, Montana State University, Bozeman, MT 59717, USA.
| | | | | | | |
Collapse
|