Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Hum Cell ; 37(4): 931-943, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38814516

ABSTRACT

Immunosuppressive regulatory cells (IRCs) play important roles in negatively regulating immune response, and are mainly divided into myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Large numbers of preclinical and clinical studies have shown that inhibition or reduction of IRCs could effectively elevate antitumor immune responses. However, several studies also reported that excessive inhibition of IRCs function is one of the main reasons causing the side effects of cancer immunotherapy. Therefore, the reasonable regulation of IRCs is crucial for improving the safety and efficiency of cancer immunotherapy. In this review, we summarised the recent research advances in the cancer immunotherapy by regulating the proportion of IRCs, and discussed the roles of IRCs in regulating tumour immune evasion and drug resistance to immunotherapies. Furthermore, we also discussed how to balance the potential opportunities and challenges of using IRCs to improve the safety of cancer immunotherapies.


Subject(s)
Immunotherapy , Myeloid-Derived Suppressor Cells , Neoplasms , T-Lymphocytes, Regulatory , Humans , Neoplasms/therapy , Neoplasms/immunology , Immunotherapy/methods , T-Lymphocytes, Regulatory/immunology , Myeloid-Derived Suppressor Cells/immunology , Tumor Escape/immunology , Drug Resistance, Neoplasm/immunology , Animals , Immunosuppression Therapy
2.
Commun Biol ; 4(1): 432, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33785850

ABSTRACT

Despite being a major bacterial factor in alerting the human immune system, the role of Staphylococcus aureus (S. aureus) lipoproteins (Lpp) in skin infections remains largely unknown. Here, we demonstrated that subcutaneous injection of S. aureus Lpp led to infiltration of neutrophils and monocytes/macrophages and induced skin lesions in mice. Lipid-moiety of S. aureus Lpp and host TLR2 was responsible for such effect. Lpp-deficient S. aureus strains exhibited smaller lesion size and reduced bacterial loads than their parental strains; the altered phenotype in bacterial loads was TLR2-independent. Lpp expression in skin infections contributed to imbalanced local hemostasis toward hypercoagulable state. Depletion of leukocytes or fibrinogen abrogated the effects induced by Lpp in terms of skin lesions and bacterial burden. Our data suggest that S. aureus Lpp induce skin inflammation and promote abscess formation that protects bacteria from innate immune killing. This suggests an intriguing bacterial immune evasion mechanism.


Subject(s)
Abscess/physiopathology , Bacterial Proteins/physiology , Lipoproteins/physiology , Staphylococcal Infections/physiopathology , Staphylococcus aureus/physiology , Abscess/microbiology , Animals , Female , Mice , Staphylococcal Infections/microbiology
3.
mBio ; 11(6)2020 11 17.
Article in English | MEDLINE | ID: mdl-33203754

ABSTRACT

Septic arthritis, one of the most dangerous joint diseases, is predominantly caused by Staphylococcus aureus In contrast, coagulase-negative staphylococci are rarely found in septic arthritis. We hypothesize that coagulases released by S. aureus, including coagulase (Coa) and von Willebrand factor-binding protein (vWbp), play potent roles in the induction of septic arthritis. Four isogenic S. aureus strains differing in expression of coagulases (wild-type [WT] Newman, Δcoa, Δvwb, and Δcoa Δvwb) were used to induce septic arthritis in both wild-type and von Willebrand factor (vWF)-deficient mice. Septic arthritis severity was greatly reduced when wild-type mice were infected with the Δcoa Δvwb and Δvwb variants compared to WT or Δcoa strains, suggesting that vWbp rather than Coa is a major virulence factor in S. aureus septic arthritis. vWF-deficient mice were more susceptible to bone damage in septic arthritis, especially when the Δvwb strain was used. Importantly, no difference in arthritis severity between the Δvwb and WT strains was observed in vWF-deficient mice. Collectively, we conclude that vWbp production by S. aureus enhances staphylococcal septic arthritis.IMPORTANCE Septic arthritis remains one of the most dangerous joint diseases with a rapidly progressive disease character. Despite advances in the use of antibiotics, permanent reductions in joint function due to joint deformation and deleterious contractures occur in up to 50% of patients with septic arthritis. So far, it is still largely unknown how S. aureus initiates and establishes joint infection. Here, we demonstrate that von Willebrand factor-binding protein expressed by S. aureus facilitates the initiation of septic arthritis. Such effect might be mediated through its interaction with a host factor (von Willebrand factor). Our finding contributes significantly to the full understanding of septic arthritis etiology and will pave the way for new therapeutic modalities for this devastating disease.


Subject(s)
Arthritis, Infectious/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , von Willebrand Factor/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins , Coagulase/genetics , Coagulase/metabolism , Female , Humans , Joints/microbiology , Mice , Mice, Inbred C57BL , Staphylococcus aureus/enzymology , Staphylococcus aureus/physiology , Virulence Factors , von Willebrand Factor/genetics
4.
Sci Rep ; 10(1): 10891, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616791

ABSTRACT

Tofacitinib, a janus kinase inhibitor, is a novel immunosuppressive drug for treatment of rheumatoid arthritis (RA). Septic arthritis (SA) and sepsis caused by Staphylococcus aureus (S. aureus), for which RA patients are at risk, are infections with high mortality. The aim of this study was to investigate the effect of tofacitinib on S. aureus infections using mouse models. In vitro tofacitinib treated mouse splenocytes were stimulated with S. aureus derived stimuli. Mice pre-treated with tofacitinib were inoculated intravenously with either arthritogenic- or septic doses of S. aureus. Arthritis severity and mortality were compared between groups. Additionally, pre-treated mice were challenged with staphylococcal toxin TSST-1 to induce shock. Tofacitinib inhibited splenocyte proliferation and IFN-γ production in response to TSST-1 and dead S. aureus. In SA, tofacitinib treatment aggravated arthritis with more severe bone erosions. However, in sepsis, treated mice displayed significantly prolonged survival compared to controls. Similarly, in staphylococcal enterotoxin-induced shock tofacitinib pre-treatment, but not late treatment dramatically reduced mortality, which was accompanied by decreased levels of TNF-α and IFN-γ. Our findings show that tofacitinib treatment increase susceptibility of SA in mice, but has a positive effect on survival in S. aureus-induced sepsis and a strong protective effect in toxin-induced shock.


Subject(s)
Arthritis, Infectious/drug therapy , Immunosuppressive Agents/therapeutic use , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Sepsis/prevention & control , Shock, Septic/prevention & control , Staphylococcal Infections/drug therapy , Animals , Arthritis, Infectious/blood , Arthritis, Infectious/chemically induced , Cytokines/blood , Disease Progression , Drug Administration Schedule , Female , Immunosuppressive Agents/toxicity , Janus Kinases/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Piperidines/toxicity , Protein Kinase Inhibitors/toxicity , Pyrimidines/toxicity , Sepsis/etiology , Shock, Septic/etiology , Spleen/drug effects , Staphylococcus aureus/drug effects , T-Lymphocytes/drug effects
5.
Sci Rep ; 10(1): 7936, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32404866

ABSTRACT

Permanent joint dysfunction is a devastating complication in patients with septic arthritis. Staphylococcus aureus (S. aureus) lipoproteins (Lpp), the predominant ligands for TLR2, are known to be arthritogenic and induce bone destruction when introduced directly into the joint. Here, we aim to investigate the importance of S. aureus Lpp and TLR2 in a hematogenous septic arthritis model, which is the most common route of infection in humans. C57BL/6 wild-type and TLR2 deficient mice were intravenously inoculated with S. aureus Newman parental strain or its lipoprotein-deficient Δlgt mutant strain. The clinical course of septic arthritis, radiological changes, and serum levels of cytokines and chemokines, were assessed. Newman strain induced more severe and frequent clinical septic polyarthritis compared to its Δlgt mutant in TLR2 deficient mice, but not in wild-type controls. Bone destruction, however, did not differ between groups. Lpp expression was associated with higher mortality, weight loss as well as impaired bacterial clearance in mouse kidneys independent of TLR2. Furthermore, Lpp expression induced increased systemic pro-inflammatory cytokine and neutrophil chemokine release. Staphylococcal Lpp are potent virulence factors in S. aureus systemic infection independent of host TLR2 signalling. However, they have a limited impact on bone erosion in hematogenous staphylococcal septic arthritis.


Subject(s)
Arthritis, Infectious/etiology , Arthritis, Infectious/pathology , Bacterial Proteins/metabolism , Hemarthrosis/etiology , Hemarthrosis/pathology , Lipoproteins/metabolism , Staphylococcal Infections/etiology , Staphylococcal Infections/pathology , Animals , Arthritis, Infectious/mortality , Bacterial Proteins/immunology , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Hemarthrosis/mortality , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Lipoproteins/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Prognosis , Severity of Illness Index , Staphylococcal Infections/mortality , Toll-Like Receptor 2/deficiency
6.
Nat Commun ; 10(1): 4333, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31551417

ABSTRACT

RNA-based therapeutics hold great promise for treating diseases and lipid nanoparticles (LNPs) represent the most advanced platform for RNA delivery. However, the fate of the LNP-mRNA after endosome-engulfing and escape from the autophagy-lysosomal pathway remains unclear. To investigate this, mRNA (encoding human erythropoietin) was delivered to cells using LNPs, which shows, for the first time, a link between LNP-mRNA endocytosis and its packaging into extracellular vesicles (endo-EVs: secreted after the endocytosis of LNP-mRNA). Endosomal escape of LNP-mRNA is dependent on the molar ratio between ionizable lipids and mRNA nucleotides. Our results show that fractions of ionizable lipids and mRNA (1:1 molar ratio of hEPO mRNA nucleotides:ionizable lipids) of endocytosed LNPs were detected in endo-EVs. Importantly, these EVs can protect the exogenous mRNA during in vivo delivery to produce human protein in mice, detected in plasma and organs. Compared to LNPs, endo-EVs cause lower expression of inflammatory cytokines.


Subject(s)
Endosomes/physiology , Erythropoietin/metabolism , Extracellular Vesicles/metabolism , Lipid Metabolism , Nanoparticles/metabolism , RNA, Messenger/metabolism , Animals , Biological Transport , Cell Line , Cytoplasm/metabolism , Endosomes/metabolism , Endosomes/ultrastructure , Erythropoietin/genetics , Female , Humans , Lipids/chemistry , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry
7.
PLoS Pathog ; 15(6): e1007877, 2019 06.
Article in English | MEDLINE | ID: mdl-31226163

ABSTRACT

Rapid bone destruction often leads to permanent joint dysfunction in patients with septic arthritis, which is mainly caused by Staphylococcus aureus (S. aureus). Staphylococcal cell wall components are known to induce joint inflammation and bone destruction. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis through TLR2. Arthritis was characterized by rapid infiltration of neutrophils and monocytes. The arthritogenic effect was mediated mainly by macrophages/monocytes and partially via TNF-α but not by neutrophils. Surprisingly, a S. aureus mutant lacking Lpp diacylglyceryl transferase (lgt) caused more severe joint inflammation, which coincided with higher bacterial loads of the lgt mutant in local joints than those of its parental strain. Coinjection of pathogenic S. aureus LS-1 with staphylococcal Lpps into mouse knee joints caused improved bacterial elimination and diminished bone erosion. The protective effect of the Lpps was mediated by their lipid moiety and was fully dependent on TLR2 and neutrophils. The blocking of CXCR2 on neutrophils resulted in total abrogation of the protective effect of the Lpps. Our data demonstrate that S. aureus Lpps elicit innate immune responses, resulting in a double-edged effect. On the one hand, staphylococcal Lpps boost septic arthritis. On the other hand, Lpps act as adjuvants and activate innate immunity, which could be useful for combating infections with multiple drug-resistant strains.


Subject(s)
Arthritis/immunology , Bacterial Proteins/immunology , Lipoproteins/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , Animals , Arthritis/genetics , Arthritis/microbiology , Arthritis/pathology , Bacterial Proteins/genetics , Female , Lipoproteins/genetics , Mice , Neutrophils/pathology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Nat Microbiol ; 3(8): 881-890, 2018 08.
Article in English | MEDLINE | ID: mdl-30013237

ABSTRACT

All bacterial infections occur within a polymicrobial environment, from which a pathogen population emerges to establish disease within a host. Emphasis has been placed on prevention of pathogen dominance by competing microflora acting as probiotics1. Here we show that the virulence of the human pathogen Staphylococcus aureus is augmented by native, polymicrobial, commensal skin flora and individual species acting as 'proinfectious agents'. The outcome is pathogen proliferation, but not commensal. Pathogenesis augmentation can be mediated by particulate cell wall peptidoglycan, reducing the S. aureus infectious dose by over 1,000-fold. This phenomenon occurs using a range of S. aureus strains and infection models and is not mediated by established receptor-mediated pathways including Nod1, Nod2, Myd88 and the NLPR3 inflammasome. During mouse sepsis, augmentation depends on liver-resident macrophages (Kupffer cells) that capture and internalize both the pathogen and the proinfectious agent, leading to reduced production of reactive oxygen species, pathogen survival and subsequent multiple liver abscess formation. The augmented infection model more closely resembles the natural situation and establishes the role of resident environmental microflora in the initiation of disease by an invading pathogen. As the human microflora is ubiquitous2, its role in increasing susceptibility to infection by S. aureus highlights potential strategies for disease prevention.


Subject(s)
Bacteria/metabolism , Peptidoglycan/pharmacology , Sepsis/microbiology , Skin/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Animals , Disease Models, Animal , Humans , Inflammasomes/metabolism , Kupffer Cells/cytology , Kupffer Cells/immunology , Macrophages/metabolism , Mice , Microbiota , Peptidoglycan/metabolism , Reactive Oxygen Species/metabolism , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/growth & development , Symbiosis , Virulence
9.
J Infect Dis ; 218(5): 791-800, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29329449

ABSTRACT

Background: Lack of receptor for advanced glycation end products (RAGE) ameliorates several infections including Staphylococcus aureus pneumonia. We sought to investigate the role of RAGE in staphylococcal skin infection in mice. Methods: Wild-type (WT) and RAGE deficient (RAGE-/-) mice were subcutaneously inoculated with S. aureus SH1000 strain in abscess-forming dose or necrotic dose. Clinical signs of dermatitis, along with histopathological changes, were compared between the groups. Results: The skin lesion size was smaller in RAGE-/- mice. Infected RAGE-/- mice expressed lower proinflammatory cytokines in local skins compared to control mice. Low dose of bacteria caused more abscess formation in RAGE-/- mice compared to skin necrosis that was more often observed in WT mice. As a result of more abscess formation, the wound healing was prolonged in RAGE-/- mice. Importantly, RAGE-/- mice had lower bacterial loads in the skin than controls, which is correlated with higher local levels of myeloperoxidase before skin infection. In vitro, enhanced phagocytic capacity of neutrophils and macrophages obtained from RAGE-/- mice compared to control mice was observed. Conclusions: RAGE deficiency up-regulates phagocytic capacity of phagocytes, resulting in lower bacterial burden in local skin and milder skin lesions in mice with staphylococcal skin infection.


Subject(s)
Abscess/pathology , Receptor for Advanced Glycation End Products/deficiency , Skin/pathology , Staphylococcal Skin Infections/pathology , Wound Healing , Abscess/genetics , Animals , Bacterial Load , Cytokines/analysis , Disease Models, Animal , Female , Histocytochemistry , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phagocytosis , Receptor for Advanced Glycation End Products/metabolism , Staphylococcal Skin Infections/genetics
10.
Infect Immun ; 85(7)2017 07.
Article in English | MEDLINE | ID: mdl-28438975

ABSTRACT

Staphylococcus aureus is a major cause of skin and soft tissue infection. The bacterium expresses four major proteases that are emerging as virulence factors: aureolysin (Aur), V8 protease (SspA), staphopain A (ScpA), and staphopain B (SspB). We hypothesized that human galectin-3, a ß-galactoside-binding lectin involved in immune regulation and antimicrobial defense, is a target for these proteases and that proteolysis of galectin-3 is a novel immune evasion mechanism. Indeed, supernatants from laboratory strains and clinical isolates of S. aureus caused galectin-3 degradation. Similar proteolytic capacities were found in Staphylococcus epidermidis isolates but not in Staphylococcus saprophyticus Galectin-3-induced activation of the neutrophil NADPH oxidase was abrogated by bacterium-derived proteolysis of galectin-3, and SspB was identified as the major protease responsible. The impact of galectin-3 and protease expression on S. aureus virulence was studied in a murine skin infection model. In galectin-3+/+ mice, SspB-expressing S. aureus caused larger lesions and resulted in higher bacterial loads than protease-lacking bacteria. No such difference in bacterial load or lesion size was detected in galectin-3-/- mice, which overall showed smaller lesion sizes than the galectin-3+/+ animals. In conclusion, the staphylococcal protease SspB inactivates galectin-3, abrogating its stimulation of oxygen radical production in human neutrophils and increasing tissue damage during skin infection.


Subject(s)
Bacterial Proteins/metabolism , Galectin 3/metabolism , Host-Pathogen Interactions , Serine Endopeptidases/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Load , Blood Proteins , Disease Models, Animal , Galectins , Humans , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Virulence
11.
PLoS One ; 12(3): e0173492, 2017.
Article in English | MEDLINE | ID: mdl-28264025

ABSTRACT

BACKGROUND: RA patients being treated with biologics are known to have an increased risk of infections. We recently demonstrated that both CTLA4 Ig and anti-TNF treatment aggravate systemic Staphylococcus aureus (S. aureus) infection in mice, but with distinct clinical manifestations. However, the effects of CTLA4 Ig and anti-TNF treatments on a local S. aureus infection (e.g., skin infection) might differ from their effects on a systemic infection. AIMS: The aim of this study was to examine the differential effects of anti-TNF versus CTLA4 Ig treatment on S. aureus skin infections in mice. METHOD: Abatacept (CTLA4 Ig), etanercept (anti-TNF treatment) or PBS was given to NMRI mice subcutaneously inoculated with S. aureus strain SH1000. The clinical signs of dermatitis, along with histopathological changes due to skin infection, were compared between the groups. RESULTS: Both CTLA4 Ig and anti-TNF treatment resulted in less severe skin infections and smaller post-infectious hyperpigmentation compared with controls. Consistent with the clinical signs of dermatitis, smaller lesion size, more epithelial hyperplasia and more granulation were found in skin biopsies from mice receiving anti-TNF compared with PBS controls. However, both CTLA4 Ig and anti-TNF therapy tended to prolong the healing time, although this finding was not statistically significant. Serum MCP-1 levels were elevated in the anti-TNF group relative to the CTLA4 Ig and PBS groups, whereas IL-6 levels were higher in PBS controls than in the other two groups. Both anti-TNF and CTLA4 Ig treatments tended to down-regulate the necrosis/apoptosis ratio in the locally infected skin tissue. Importantly, no tangible difference was found in the bacterial burden among groups. CONCLUSION: Both CTLA4 Ig and anti-TNF therapies attenuate disease severity but may prolong the healing time required for S. aureus skin infections. Neither treatment has an impact on bacterial clearance in skin tissues.


Subject(s)
Abatacept/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Dermatitis/microbiology , Etanercept/pharmacology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Apoptosis , Bacterial Load , Cytokines/blood , Cytokines/metabolism , Dermatitis/drug therapy , Dermatitis/immunology , Dermatitis/metabolism , Disease Models, Animal , Female , Hyperpigmentation , Immunosuppressive Agents/pharmacology , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Necrosis , Phenotype , Severity of Illness Index , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology
12.
PLoS One ; 12(2): e0171222, 2017.
Article in English | MEDLINE | ID: mdl-28152087

ABSTRACT

BACKGROUND: Permanent joint dysfunction due to bone destruction occurs in up to 50% of patients with septic arthritis. Recently, imaging technologies such as micro computed tomography (µCT) scan have been widely used for preclinical models of autoimmune joint disorders. However, the radiological features of septic arthritis in mice are still largely unknown. METHODS: NMRI mice were intravenously or intra-articularly inoculated with S. aureus Newman or LS-1 strain. The radiological and clinical signs of septic arthritis were followed for 10 days using µCT. We assessed the correlations between joint radiological changes and clinical signs, histological changes, and serum levels of cytokines. RESULTS: On days 5-7 after intravenous infection, bone destruction verified by µCT became evident in most of the infected joints. Radiological signs of bone destruction were dependent on the bacterial dose. The site most commonly affected by septic arthritis was the distal femur in knees. The bone destruction detected by µCT was positively correlated with histological changes in both local and hematogenous septic arthritis. The serum levels of IL-6 were significantly correlated with the severity of joint destruction. CONCLUSION: µCT is a sensitive method for monitoring disease progression and determining the severity of bone destruction in a mouse model of septic arthritis. IL-6 may be used as a biomarker for bone destruction in septic arthritis.


Subject(s)
Arthritis, Infectious/diagnostic imaging , Staphylococcal Infections/diagnostic imaging , Animals , Arthritis, Infectious/pathology , Disease Models, Animal , Disease Progression , Female , Joints/diagnostic imaging , Joints/microbiology , Joints/pathology , Mice , Staphylococcal Infections/pathology , X-Ray Microtomography
13.
PLoS One ; 11(12): e0167287, 2016.
Article in English | MEDLINE | ID: mdl-27907047

ABSTRACT

BACKGROUND: Septic arthritis is a serious joint disease often caused by Staphylococcus aureus (S. aureus). Receptor for Advanced Glycation End products (RAGE) has an important role in several infections. We sought to investigate the role of RAGE in staphylococcal septic arthritis and sepsis in mice. METHODS: Wild-type (WT) and RAGE deficient (RAGE-/-) mice were intra-articularly or intravenously inoculated with an arthritic or septic dose of S. aureus LS-1 strain. Clinical arthritis, weight development and mortality were monitored for 14 days. Serum levels of cytokines, kidney bacterial loads as well as micro-CT and histopathology of the joints were assessed. RESULTS: RAGE-/- mice with septic arthritis had significantly lower IL-17A and higher bone mineral density (BMD) compared to the control group. However, no significant differences between the groups were observed regarding the weight loss, the severity and frequency of arthritis, and bacterial loads in the kidneys. In mice with sepsis, the overall mortality rate was similar in RAGE-/- (39%) and in WT mice (45%). However, RAGE-/- mice with sepsis had significantly higher bacterial load in their kidneys compared to the WT controls. In line with data from hematogenous S. aureus arthritis, RAGE deficiency had no impact on arthritis severity in local joint infection. CONCLUSIONS: Our results indicate that lack of RAGE has no significant impact on septic arthritis. However, RAGE-/- mice had significantly higher BMD compared to WT mice, which coincided with lower IL-17A in RAGE-/- mice. In sepsis, RAGE deficiency impairs bacterial kidney clearance.


Subject(s)
Arthritis, Infectious/genetics , Arthritis, Infectious/microbiology , Receptor for Advanced Glycation End Products/deficiency , Sepsis/genetics , Sepsis/microbiology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Animals , Arthritis, Infectious/blood , Arthritis, Infectious/diagnosis , Bacterial Load , Biomarkers , Cytokines/blood , Disease Models, Animal , Female , Inflammation Mediators/blood , Knee Joint/diagnostic imaging , Knee Joint/microbiology , Knee Joint/pathology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Sepsis/blood , Sepsis/diagnosis , Severity of Illness Index , Staphylococcal Infections/blood , Staphylococcal Infections/diagnosis , X-Ray Microtomography
14.
Infect Immun ; 84(4): 930-939, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787717

ABSTRACT

The complement system plays an essential role in the innate immune response and protection against bacterial infections. However, detailed knowledge regarding the role of complement in Staphylococcus aureus septic arthritis is still largely missing. In this study, we elucidated the roles of selected complement proteins in S. aureus septic arthritis. Mice lacking the complement component 3 (C3(-/-)), complement factor B (fB(-/-)), and receptor for C3-derived anaphylatoxin C3a (C3aR(-/-)) and wild-type (WT) control mice were intravenously or intra-articularly inoculated with S. aureus strain Newman. The clinical course of septic arthritis, as well as histopathological and radiological changes in joints, was assessed. After intravenous inoculation, arthritis severity and frequency were significantly higher in C3(-/-)mice than in WT controls, whereas fB(-/-)mice displayed intermediate arthritis severity and frequency. This was in accordance with both histopathological and radiological findings. C3, but not fB, deficiency was associated with greater weight loss, more frequent kidney abscesses, and higher bacterial burden in kidneys. S. aureus opsonized with C3(-/-)sera displayed decreased uptake by mouse peritoneal macrophages compared with bacteria opsonized with WT or fB(-/-)sera. C3aR deficiency had no effect on the course of hematogenous S. aureus septic arthritis. We conclude that C3 deficiency increases susceptibility to hematogenous S. aureus septic arthritis and impairs host bacterial clearance, conceivably due to diminished opsonization and phagocytosis of S. aureus.


Subject(s)
Arthritis, Infectious/immunology , Complement C3/metabolism , Complement Factor B/metabolism , Receptors, G-Protein-Coupled/metabolism , Staphylococcal Infections/immunology , Animals , Arthritis, Infectious/genetics , Complement C3/genetics , Complement Factor B/genetics , Gene Expression Regulation/physiology , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Phagocytosis/physiology , Receptors, G-Protein-Coupled/genetics , Staphylococcal Infections/pathology , Staphylococcus aureus
15.
Appl Environ Microbiol ; 82(1): 394-401, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26519394

ABSTRACT

Staphylococcus aureus biofilm infections of indwelling medical devices are a major medical challenge because of their high prevalence and antibiotic resistance. As fibrin plays an important role in S. aureus biofilm formation, we hypothesize that coating of the implant surface with fibrinolytic agents can be used as a new method of antibiofilm prophylaxis. The effect of tissue plasminogen activator (tPA) coating on S. aureus biofilm formation was tested with in vitro microplate biofilm assays and an in vivo mouse model of biofilm infection. tPA coating efficiently inhibited biofilm formation by various S. aureus strains. The effect was dependent on plasminogen activation by tPA, leading to subsequent local fibrin cleavage. A tPA coating on implant surfaces prevented both early adhesion and later biomass accumulation. Furthermore, tPA coating increased the susceptibility of biofilm infections to antibiotics. In vivo, significantly fewer bacteria were detected on the surfaces of implants coated with tPA than on control implants from mice treated with cloxacillin. Fibrinolytic coatings (e.g., with tPA) reduce S. aureus biofilm formation both in vitro and in vivo, suggesting a novel way to prevent bacterial biofilm infections of indwelling medical devices.


Subject(s)
Biofilms/growth & development , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Tissue Plasminogen Activator/pharmacology , Animals , Biofilms/drug effects , Cloxacillin/administration & dosage , Disease Models, Animal , Fibrin/metabolism , Humans , In Vitro Techniques , Mice , Staphylococcus aureus/drug effects , Surface Properties
16.
J Infect Dis ; 213(1): 139-48, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26136471

ABSTRACT

Staphylococcus aureus biofilms, a leading cause of persistent infections, are highly resistant to immune defenses and antimicrobial therapies. In the present study, we investigated the contribution of fibrin and staphylokinase (Sak) to biofilm formation. In both clinical S. aureus isolates and laboratory strains, high Sak-producing strains formed less biofilm than strains that lacked Sak, suggesting that Sak prevents biofilm formation. In addition, Sak induced detachment of mature biofilms. This effect depended on plasminogen activation by Sak. Host-derived fibrin, the main substrate cleaved by Sak-activated plasminogen, was a major component of biofilm matrix, and dissolution of this fibrin scaffold greatly increased susceptibility of biofilms to antibiotics and neutrophil phagocytosis. Sak also attenuated biofilm-associated catheter infections in mouse models. In conclusion, our results reveal a novel role for Sak-induced plasminogen activation that prevents S. aureus biofilm formation and induces detachment of existing biofilms through proteolytic cleavage of biofilm matrix components.


Subject(s)
Biofilms/drug effects , Metalloendopeptidases/metabolism , Staphylococcus aureus/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Female , Fibrin/metabolism , Male , Metalloendopeptidases/pharmacology , Mice , Mice, Inbred C57BL , Plasminogen/metabolism , Staphylococcus aureus/drug effects
17.
PLoS One ; 10(7): e0131645, 2015.
Article in English | MEDLINE | ID: mdl-26135738

ABSTRACT

BACKGROUND: Interleukin-1 receptor antagonist (IL-1Ra) is the primary therapy against autoinflammatory syndromes with robust efficacy in reducing systemic inflammation and associated organ injury. However, patients receiving IL-1Ra might be at increased risk of acquiring serious infections. AIMS: To study whether IL-1Ra treatment deteriorates Staphylococcus aureus (S. aureus) septic arthritis and sepsis in mice. METHOD: NMRI mice were treated with anakinra (IL-1Ra) daily for 7 days before intravenous inoculation with S. aureus strain Newman in both arthritogenic and lethal doses. The clinical course of septic arthritis, histopathological and radiological changes of the joints, as well as the mortality were compared between IL-1Ra treated and control groups. RESULTS: IL-1Ra treated mice developed more frequent and severe clinical septic arthritis. Also, the frequency of polyarthritis was significantly higher in the mice receiving IL-1Ra therapy. In line with the data from clinical arthritis, both histological and radiological signs of septic arthritis were more pronounced in IL-1Ra treated group compared to controls. Importantly, the mortality of IL-1Ra treated mice was significantly higher than PBS treated controls. CONCLUSION: IL-1Ra treatment significantly aggravated S. aureus induced septic arthritis and increased the mortality in these mice.


Subject(s)
Arthritis, Infectious/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Sepsis/drug therapy , Staphylococcal Infections/drug therapy , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/microbiology , Arthritis, Infectious/microbiology , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/metabolism , Joints/pathology , Kidney/pathology , Macrophages, Peritoneal/metabolism , Mice , Sepsis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus , Staphylococcus aureus , X-Ray Microtomography
18.
J Infect Dis ; 212(8): 1308-16, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-25838268

ABSTRACT

BACKGROUND: The development of biologics has greatly increased the quality of life and the life expectancy of many patients with rheumatoid arthritis. However, a large number of these patients have an increased risk of developing serious infections. The aim of this study was to examine differential effects of anti-tumor necrosis factor (TNF) treatment and CTLA4 immunoglobulin (Ig) treatment on both immunological response and host defense in a murine model of septic arthritis. METHODS: Abatacept (CTLA4-Ig), etanercept (anti-TNF), or phosphate-buffered saline were given to NMRI mice intravenously inoculated with Staphylococcus aureus. The clinical course of septic arthritis and histopathological and radiological changes of joints were compared among the groups. RESULTS: Mice receiving CTLA4-Ig treatment had more-severe septic arthritis, compared with controls and mice receiving anti-TNF treatment. Anti-TNF treatment led to more-severe weight loss and kidney abscesses, as well as a higher bacterial burden in the kidneys. Mice receiving CTLA4-Ig therapy had lower serum levels of interleukin 4, whereas mice receiving anti-TNF therapy had higher levels of TNF-α. Both iNOS and arginase-1 expression were reduced in peritoneal macrophages from mice receiving CTLA4-Ig, compared with expression in the anti-TNF group. CONCLUSIONS: CTLA4-Ig therapy significantly increased the susceptibility to S. aureus septic arthritis in mice, whereas anti-TNF therapy deteriorated host bacterial clearance, resulting in more-severe weight loss and kidney abscesses.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Infectious/immunology , Arthritis, Rheumatoid/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Abatacept/therapeutic use , Animals , Arthritis, Infectious/microbiology , Arthritis, Rheumatoid/microbiology , Etanercept/therapeutic use , Female , Joints/immunology , Joints/pathology , Mice , Staphylococcal Infections/microbiology , Tumor Necrosis Factor-alpha/immunology
19.
Virology ; 456-457: 227-37, 2014 May.
Article in English | MEDLINE | ID: mdl-24889242

ABSTRACT

Polycomb ring finger oncogene BMI1 (B cell-specific Moloney murine leukemia virus integration site 1) plays a critical role in development of several types of cancers. Here, we report an inverse relationship between levels of BMI1 expression and adenovirus (Ad) progeny production. Enforced BMI1 expression in A549 cells impaired Ad progeny production. In contrast, knocking-down of endogenous BMI1 expression enhanced progeny production of a conditionally replicating Ad and wild-type Ad5 and Ad11p. Ad vectors overexpressing BMI1 were not impaired in the replication of progeny genomes and in the expression of E1A and Ad structural proteins. However, 293 cells infected by Ad vector overexpressing BMI1 contained a large proportion of morphologically irregular Ad particles. This effect was reversed in 293 cells pre-treated with the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) in parallel with the production of infectious Ad particles. Our findings suggest an inhibitory role of BMI1 in Ad morphogenesis that can be implied in Ad tropism and Ad-mediated cancer therapy.


Subject(s)
Adenoviridae/immunology , Adenoviridae/physiology , Histone Deacetylases/metabolism , Host-Pathogen Interactions , Polycomb Repressive Complex 1/metabolism , Viral Tropism , Virus Assembly , Cell Line , Humans
20.
PLoS One ; 9(1): e87281, 2014.
Article in English | MEDLINE | ID: mdl-24489888

ABSTRACT

Increased PDGFRA signaling is an essential pathogenic factor in many subtypes of gliomas. In this context the cell surface expression of PDGFRA is an important determinant of ligand sensing in the glioma microenvironment. However, the regulation of spatial distribution of PDGFRA in glioma cells remains poorly characterized. Here, we report that cell surface PDGFRA expression in gliomas is negatively regulated by an ERK-dependent mechanism, resulting in reduced proliferation of glioma cells. Glioma tumor tissues and their corresponding cell lines were isolated from 14 patients and analyzed by single-cell imaging and flow cytometry. In both cell lines and their corresponding tumor samples, glioma cell proliferation correlated with the extent of surface expression of PDGFRA. High levels of surface PDGFRA also correlated to high tubulin expression in glioma tumor tissue in vivo. In glioma cell lines, surface PDGFRA declined following treatment with inhibitors of tubulin, actin and dynamin. Screening of a panel of small molecule compounds identified the MEK inhibitor U0126 as a potent inhibitor of surface PDGFRA expression. Importantly, U0126 inhibited surface expression in a reversible, dose- and time-dependent manner, without affecting general PDGFRA expression. Treatment with U0126 resulted in reduced co-localization between PDGFRA and intracellular trafficking molecules e.g. clathrin, RAB11 and early endosomal antigen-1, in parallel with enhanced co-localization between PDGFRA and the Golgi cisternae maker, Giantin, suggesting a deviation of PDGFRA from the endosomal trafficking and recycling compartment, to the Golgi network. Furthermore, U0126 treatment in glioma cells induced an initial inhibition of ERK1/2 phosphorylation, followed by up-regulated ERK1/2 phosphorylation concomitant with diminished surface expression of PDGFRA. Finally, down-regulation of surface PDGFRA expression by U0126 is concordant with reduced glioma cell proliferation. These findings suggest that manipulation of spatial expression of PDGFRA can potentially be used to combat gliomas.


Subject(s)
Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Butadienes/pharmacology , Cell Membrane/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Transport , Tumor Cells, Cultured , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL