Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Thorax ; 79(8): 778-787, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38508718

ABSTRACT

INTRODUCTION: Novel therapeutic strategies are urgently needed for Mycobacterium avium complex pulmonary disease (MAC-PD). Human mesenchymal stromal cells (MSCs) can directly inhibit MAC growth, but their effect on intracellular bacilli is unknown. We investigated the ability of human MSCs to reduce bacterial replication and inflammation in MAC-infected macrophages and in a murine model of MAC-PD. METHODS: Human monocyte-derived macrophages (MDMs) were infected with M. avium Chester strain and treated with human bone marrow-derived MSCs. Intracellular and extracellular colony-forming units (CFUs) were counted at 72 hours. Six-week-old female balb/c mice were infected by nebulisation of M. avium Chester. Mice were treated with 1×106 intravenous human MSCs or saline control at 21 and 28 days post-infection. Lungs, liver and spleen were harvested 42 days post-infection for bacterial counts. Cytokines were quantified by ELISA. RESULTS: MSCs reduced intracellular bacteria in MDMs over 72 hours (median 35% reduction, p=0.027). MSC treatment increased extracellular concentrations of prostaglandin E2 (PGE2) (median 10.1-fold rise, p=0.002) and reduced tumour necrosis factor-α (median 28% reduction, p=0.025). Blocking MSC PGE2 production by cyclo-oxygenase-2 (COX-2) inhibition with celecoxib abrogated the antimicrobial effect, while this was restored by adding exogenous PGE2. MSC-treated mice had lower pulmonary CFUs (median 18% reduction, p=0.012), but no significant change in spleen or liver CFUs compared with controls. CONCLUSION: MSCs can modulate inflammation and reduce intracellular M. avium growth in human macrophages via COX-2/PGE2 signalling and inhibit pulmonary bacterial replication in a murine model of chronic MAC-PD.


Subject(s)
Disease Models, Animal , Mesenchymal Stem Cells , Mice, Inbred BALB C , Mycobacterium avium-intracellulare Infection , Animals , Mice , Female , Humans , Mycobacterium avium-intracellulare Infection/microbiology , Mycobacterium avium Complex , Mesenchymal Stem Cell Transplantation/methods , Macrophages/microbiology , Dinoprostone/metabolism , Sulfonamides/pharmacology , Mycobacterium avium
2.
Proc Natl Acad Sci U S A ; 117(30): 18018-18028, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32651278

ABSTRACT

CCN3 is a matricellular protein that promotes oligodendrocyte progenitor cell differentiation and myelination in vitro and ex vivo. CCN3 is therefore a candidate of interest in central nervous system (CNS) myelination and remyelination, and we sought to investigate the expression and role of CCN3 during these processes. We found CCN3 to be expressed predominantly by neurons in distinct areas of the CNS, primarily the cerebral cortex, hippocampus, amygdala, suprachiasmatic nuclei, anterior olfactory nuclei, and spinal cord gray matter. CCN3 was transiently up-regulated following demyelination in the brain of cuprizone-fed mice and spinal cord lesions of mice injected with lysolecithin. However, CCN3-/- mice did not exhibit significantly different numbers of oligodendroglia or differentiated oligodendrocytes in the healthy or remyelinating CNS, compared to WT controls. These results suggest that despite robust and dynamic expression in the CNS, CCN3 is not required for efficient myelination or remyelination in the murine CNS in vivo.


Subject(s)
Central Nervous System/metabolism , Demyelinating Diseases/etiology , Gene Expression Regulation , Nephroblastoma Overexpressed Protein/genetics , Remyelination/genetics , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Fluorescent Antibody Technique , Mice , Myelin Sheath/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology
3.
Mediators Inflamm ; 2021: 6682657, 2021.
Article in English | MEDLINE | ID: mdl-33828414

ABSTRACT

BACKGROUND: Elevated levels of the cysteine protease cathepsin S (CatS) are associated with chronic mucoobstructive lung diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). We have previously demonstrated that prophylactic treatment with a CatS inhibitor from birth reduces inflammation, mucus plugging, and lung tissue damage in juvenile ß-epithelial Na+ channel-overexpressing transgenic (ßENaC-Tg) mice with chronic inflammatory mucoobstructive lung disease. In this study, we build upon this work to examine the effects of therapeutic intervention with a CatS inhibitor in adult ßENaC-Tg mice with established disease. METHODS: ßENaC-Tg mice and wild-type (WT) littermates were treated with a CatS inhibitor from 4 to 6 weeks of age, and CatS-/- ßENaC-Tg mice were analysed at 6 weeks of age. Bronchoalveolar lavage (BAL) fluid inflammatory cell counts were quantified, and lung tissue destruction and mucus obstruction were analysed histologically. RESULTS: At 6 weeks of age, ßENaC-Tg mice developed significant airway inflammation, lung tissue damage, and mucus plugging when compared to WT mice. CatS-/- ßENaC-Tg mice and ßENaC-Tg mice receiving inhibitor had significantly reduced airway mononuclear and polymorphonuclear (PMN) cell counts as well as mucus plugging. However, in contrast to CatS-/- ßENaC-Tg mice, therapeutic inhibition of CatS in ßENaC-Tg mice had no effect on established emphysema-like lung tissue damage. CONCLUSIONS: These results suggest that while early CatS targeting may be required to prevent the onset and progression of lung tissue damage, therapeutic CatS targeting effectively inhibited airway inflammation and mucus obstruction. These results indicate the important role CatS may play in the pathogenesis and progression of mucoobstructive lung disease.


Subject(s)
Cathepsins/antagonists & inhibitors , Cystic Fibrosis , Epithelial Sodium Channels , Animals , Cystic Fibrosis/pathology , Disease Models, Animal , Inflammation/drug therapy , Inflammation/pathology , Lung/pathology , Mice , Mice, Transgenic , Mucus
4.
Article in English | MEDLINE | ID: mdl-30858214

ABSTRACT

Using a murine model of Klebsiella pneumoniae bacterial infection, we demonstrate that gentamicin dissolving microarray patches, applied to murine ears, could control K. pneumoniae infection. Mice treated with microarray patches had reduced bacterial burden in the nasal-associated lymphoid tissue and lungs compared with their untreated counterparts. This proof of concept study represents the first published data on the in vivo delivery of the antibiotic gentamicin via dissolving microarray patches, resulting in the control of bacterial infection.


Subject(s)
Gentamicins/therapeutic use , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/pathogenicity , Animals , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Mice
5.
PLoS Pathog ; 13(11): e1006696, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29112952

ABSTRACT

Klebsiella pneumoniae is a significant cause of nosocomial pneumonia and an alarming pathogen owing to the recent isolation of multidrug resistant strains. Understanding of immune responses orchestrating K. pneumoniae clearance by the host is of utmost importance. Here we show that type I interferon (IFN) signaling protects against lung infection with K. pneumoniae by launching bacterial growth-controlling interactions between alveolar macrophages and natural killer (NK) cells. Type I IFNs are important but disparate and incompletely understood regulators of defense against bacterial infections. Type I IFN receptor 1 (Ifnar1)-deficient mice infected with K. pneumoniae failed to activate NK cell-derived IFN-γ production. IFN-γ was required for bactericidal action and the production of the NK cell response-amplifying IL-12 and CXCL10 by alveolar macrophages. Bacterial clearance and NK cell IFN-γ were rescued in Ifnar1-deficient hosts by Ifnar1-proficient NK cells. Consistently, type I IFN signaling in myeloid cells including alveolar macrophages, monocytes and neutrophils was dispensable for host defense and IFN-γ activation. The failure of Ifnar1-deficient hosts to initiate a defense-promoting crosstalk between alveolar macrophages and NK cell was circumvented by administration of exogenous IFN-γ which restored endogenous IFN-γ production and restricted bacterial growth. These data identify NK cell-intrinsic type I IFN signaling as essential driver of K. pneumoniae clearance, and reveal specific targets for future therapeutic exploitations.


Subject(s)
Interferon Type I/immunology , Killer Cells, Natural/immunology , Klebsiella Infections/immunology , Macrophages, Alveolar/immunology , Signal Transduction/immunology , Animals , Drug Resistance, Multiple/immunology , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor Cross-Talk/immunology , Respiratory Tract Infections/immunology
6.
J Antimicrob Chemother ; 73(12): 3391-3397, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30219825

ABSTRACT

Objectives: To determine the antimicrobial activity of ALX-009, a combination of bovine lactoferrin and hypothiocyanite, in sputum against Pseudomonas aeruginosa and Burkholderia cepacia complex (Bcc), key pathogens causing infection in the lungs of cystic fibrosis (CF) patients. Methods: The antimicrobial activity of ALX-009 against clinical respiratory P. aeruginosa isolates was determined by time-kill assay. Sputum from CF patients was treated with ALX-009, either alone or in combination with tobramycin, and the effect on P. aeruginosa, Bcc and total sputum density was determined. Results: Time-kill assay indicated that ALX-009 was bactericidal at 24 h against 4/4 P. aeruginosa isolates under aerobic conditions, and against 3/4 isolates under anaerobic conditions. ALX-009 was also bactericidal against P. aeruginosa in sputum samples at 6 h (n = 22/24 samples) and 24 h (n = 14/24 samples), and demonstrated significantly greater activity than tobramycin at both timepoints. Activity against Bcc in sputum samples (n = 9) was also demonstrated, but the magnitude of change in Bcc density was less than for P. aeruginosa. To determine the effect of treating sputum with two doses of ALX-009, similar to current regimens for inhaled antibiotics, aliquots of a further 10 sputum samples positive for P. aeruginosa were treated with one (t = 0 h) or two doses (t = 0 h, t = 12 h) of ALX-009; treatment with two doses resulted in bactericidal activity in 7/10 samples at 34 h compared with only 3/10 samples when treatment was with one dose. Conclusions: ALX-009 demonstrates promise as a novel antimicrobial that could be used to decrease P. aeruginosa density in the lungs of people with CF.


Subject(s)
Anti-Infective Agents/pharmacology , Burkholderia cepacia complex/drug effects , Cystic Fibrosis/microbiology , Lactoferrin/pharmacology , Pseudomonas aeruginosa/drug effects , Sputum/microbiology , Thiocyanates/pharmacology , Humans , Microbial Sensitivity Tests , Microbial Viability/drug effects
8.
PLoS Pathog ; 11(1): e1004627, 2015 01.
Article in English | MEDLINE | ID: mdl-25633080

ABSTRACT

Klebsiella pneumoniae is a significant human pathogen, in part due to high rates of multidrug resistance. RamA is an intrinsic regulator in K. pneumoniae established to be important for the bacterial response to antimicrobial challenge; however, little is known about its possible wider regulatory role in this organism during infection. In this work, we demonstrate that RamA is a global transcriptional regulator that significantly perturbs the transcriptional landscape of K. pneumoniae, resulting in altered microbe-drug or microbe-host response. This is largely due to the direct regulation of 68 genes associated with a myriad of cellular functions. Importantly, RamA directly binds and activates the lpxC, lpxL-2 and lpxO genes associated with lipid A biosynthesis, thus resulting in modifications within the lipid A moiety of the lipopolysaccharide. RamA-mediated alterations decrease susceptibility to colistin E, polymyxin B and human cationic antimicrobial peptide LL-37. Increased RamA levels reduce K. pneumoniae adhesion and uptake into macrophages, which is supported by in vivo infection studies, that demonstrate increased systemic dissemination of ramA overexpressing K. pneumoniae. These data establish that RamA-mediated regulation directly perturbs microbial surface properties, including lipid A biosynthesis, which facilitate evasion from the innate host response. This highlights RamA as a global regulator that confers pathoadaptive phenotypes with implications for our understanding of the pathogenesis of Enterobacter, Salmonella and Citrobacter spp. that express orthologous RamA proteins.


Subject(s)
Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Host-Pathogen Interactions/genetics , Klebsiella pneumoniae/genetics , Lipopolysaccharides/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Base Sequence , Cells, Cultured , Gene Expression Regulation, Bacterial , Genes, Bacterial , Klebsiella Infections/genetics , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymyxins/pharmacology , Regulon
9.
J Immunol ; 195(1): 87-95, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002979

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a devastating disorder characterized by increased alveolar permeability with no effective treatment beyond supportive care. Current mechanisms underlying ARDS focus on alveolar endothelial and epithelial injury caused by products of innate immune cells and platelets. However, the role of adaptive immune cells in ARDS remains largely unknown. In this study, we report that expansion of Ag-specific αßTh17 cells contributes to ARDS by local secretion of IL-17A, which in turn directly increases alveolar epithelial permeability. Mice with a highly restrictive defect in Ag-specific αßTh17 cells were protected from experimental ARDS induced by a single dose of endotracheal LPS. Loss of IL-17 receptor C or Ab blockade of IL-17A was similarly protective, further suggesting that IL-17A released by these cells was responsible for this effect. LPS induced a rapid and specific clonal expansion of αßTh17 cells in the lung, as determined by deep sequencing of the hypervariable CD3RßVJ region of the TCR. Our findings could be relevant to ARDS in humans, because we found significant elevation of IL-17A in bronchoalveolar lavage fluid from patients with ARDS, and rIL-17A directly increased permeability across cultured human alveolar epithelial monolayers. These results reveal a previously unexpected role for adaptive immune responses that increase alveolar permeability in ARDS and suggest that αßTh17 cells and IL-17A could be novel therapeutic targets for this currently untreatable disease.


Subject(s)
Interleukin-17/immunology , Pulmonary Alveoli/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Respiratory Distress Syndrome/immunology , Th17 Cells/immunology , Adaptive Immunity , Animals , Antibodies/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Interleukin-17/antagonists & inhibitors , Interleukin-17/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Permeability , Primary Cell Culture , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Rats , Rats, Sprague-Dawley , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Th17 Cells/drug effects , Th17 Cells/pathology
10.
Am J Respir Crit Care Med ; 193(4): 407-16, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26488187

ABSTRACT

RATIONALE: IL-17A is purported to help drive early pathogenesis in acute respiratory distress syndrome (ARDS) by enhancing neutrophil recruitment. Although IL-17A is the archetypal cytokine of T-helper 17 cells, it is produced by a number of lymphocytes, the source during ARDS being unknown. OBJECTIVES: To identify the cellular source and the role of IL-17A in the early phase of lung injury. METHODS: Lung injury was induced in wild-type (C57BL/6) and IL-17 knockout (KO) mice with aerosolized LPS (100 µg) or Pseudomonas aeruginosa infection. Detailed phenotyping of the cells expressing RORγt, the transcriptional regulator of IL-17 production, in the mouse lung at 24 hours was performed by flow cytometry. MEASUREMENTS AND MAIN RESULTS: A 100-fold reduction in neutrophil infiltration was observed in the lungs of the IL-17A KO compared with wild-type mice. The majority of RORγt(+) cells in the mouse lung were the recently identified group 3 innate lymphoid cells (ILC3s). Detailed characterization revealed these pulmonary ILC3s (pILC3s) to be discrete from those described in the gut. The critical role of these cells was verified by inducing injury in recombinase-activating gene 2 KO mice, which lack T cells but retain innate lymphoid cells. No amelioration of pathology was observed in the recombinase-activating gene 2 KO mice. CONCLUSIONS: IL-17 is rapidly produced during lung injury and significantly contributes to early immunopathogenesis. This is orchestrated largely by a distinct population of pILC3s. Modulation of the activity of pILC3s may potentiate early control of the inflammatory dysregulation seen in ARDS, opening up new therapeutic targets.


Subject(s)
Interleukin-17/biosynthesis , Lymphocytes/pathology , Respiratory Distress Syndrome/pathology , Animals , Disease Models, Animal , Female , Flow Cytometry , Lung/pathology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Respiratory Distress Syndrome/metabolism
11.
PLoS Pathog ; 10(5): e1004085, 2014 May.
Article in English | MEDLINE | ID: mdl-24788397

ABSTRACT

Bacillus anthracis produces a binary toxin composed of protective antigen (PA) and one of two subunits, lethal factor (LF) or edema factor (EF). Most studies have concentrated on induction of toxin-specific antibodies as the correlate of protective immunity, in contrast to which understanding of cellular immunity to these toxins and its impact on infection is limited. We characterized CD4+ T cell immunity to LF in a panel of humanized HLA-DR and DQ transgenic mice and in naturally exposed patients. As the variation in antigen presentation governed by HLA polymorphism has a major impact on protective immunity to specific epitopes, we examined relative binding affinities of LF peptides to purified HLA class II molecules, identifying those regions likely to be of broad applicability to human immune studies through their ability to bind multiple alleles. Transgenics differing only in their expression of human HLA class II alleles showed a marked hierarchy of immunity to LF. Immunogenicity in HLA transgenics was primarily restricted to epitopes from domains II and IV of LF and promiscuous, dominant epitopes, common to all HLA types, were identified in domain II. The relevance of this model was further demonstrated by the fact that a number of the immunodominant epitopes identified in mice were recognized by T cells from humans previously infected with cutaneous anthrax and from vaccinated individuals. The ability of the identified epitopes to confer protective immunity was demonstrated by lethal anthrax challenge of HLA transgenic mice immunized with a peptide subunit vaccine comprising the immunodominant epitopes that we identified.


Subject(s)
Anthrax Vaccines , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , CD4-Positive T-Lymphocytes/immunology , HLA Antigens/genetics , Immunity, Cellular/genetics , Polymorphism, Genetic , Skin Diseases, Bacterial/prevention & control , Adult , Amino Acid Sequence , Animals , Anthrax/immunology , Anthrax Vaccines/chemistry , Anthrax Vaccines/therapeutic use , Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Epitope Mapping , HLA Antigens/immunology , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Molecular Targeted Therapy , Skin Diseases, Bacterial/immunology , Young Adult
12.
Am J Respir Crit Care Med ; 189(12): 1520-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24716610

ABSTRACT

RATIONALE: Increasing epithelial repair and regeneration may hasten resolution of lung injury in patients with the acute respiratory distress syndrome (ARDS). In animal models of ARDS, keratinocyte growth factor (KGF) reduces injury and increases epithelial proliferation and repair. The effect of KGF in the human alveolus is unknown. OBJECTIVES: To test whether KGF can attenuate alveolar injury in a human model of ARDS. METHODS: Volunteers were randomized to intravenous KGF (60 µg/kg) or placebo for 3 days, before inhaling 50 µg LPS. Six hours later, subjects underwent bronchoalveolar lavage (BAL) to quantify markers of alveolar inflammation and cell-specific injury. MEASUREMENTS AND MAIN RESULTS: KGF did not alter leukocyte infiltration or markers of permeability in response to LPS. KGF increased BAL concentrations of surfactant protein D, matrix metalloproteinase (MMP)-9, IL-1Ra, granulocyte-macrophage colony-stimulating factor (GM-CSF), and C-reactive protein. In vitro, BAL fluid from KGF-treated subjects inhibited pulmonary fibroblast proliferation, but increased alveolar epithelial proliferation. Active MMP-9 increased alveolar epithelial wound repair. Finally, BAL from the KGF-pretreated group enhanced macrophage phagocytic uptake of apoptotic epithelial cells and bacteria compared with BAL from the placebo-treated group. This effect was blocked by inhibiting activation of the GM-CSF receptor. CONCLUSIONS: KGF treatment increases BAL surfactant protein D, a marker of type II alveolar epithelial cell proliferation in a human model of acute lung injury. Additionally, KGF increases alveolar concentrations of the antiinflammatory cytokine IL-1Ra, and mediators that drive epithelial repair (MMP-9) and enhance macrophage clearance of dead cells and bacteria (GM-CSF). Clinical trial registered with ISRCTN 98813895.


Subject(s)
Acute Lung Injury/drug therapy , Epithelial Cells/drug effects , Fibroblast Growth Factor 7/therapeutic use , Models, Biological , Protective Agents/therapeutic use , Pulmonary Alveoli/drug effects , Respiratory Distress Syndrome/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Administration, Intravenous , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Bronchoalveolar Lavage , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Administration Schedule , Epithelial Cells/physiology , Female , Fibroblast Growth Factor 7/pharmacology , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Lipopolysaccharides , Male , Middle Aged , Protective Agents/pharmacology , Pulmonary Alveoli/physiology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/prevention & control , Wound Healing/drug effects , Young Adult
13.
Nat Commun ; 15(1): 1870, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38467607

ABSTRACT

Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.


Subject(s)
Remyelination , Humans , Male , Female , Mice , Animals , Aged , Remyelination/physiology , T-Lymphocytes, Regulatory/metabolism , Oligodendroglia/physiology , Cell Differentiation/physiology , Myelin Sheath/metabolism , Aging , Central Nervous System
14.
Acta Neuropathol ; 126(4): 501-15, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934116

ABSTRACT

Multiple sclerosis is considered a disease of complex autoimmune etiology, yet there remains a lack of consensus as to specific immune effector mechanisms. Recent analyses of experimental autoimmune encephalomyelitis, the common mouse model of multiple sclerosis, have investigated the relative contribution of Th1 and Th17 CD4 T cell subsets to initial autoimmune central nervous system (CNS) damage. However, inherent in these studies are biases influenced by the adjuvant and toxin needed to break self-tolerance. We investigated spontaneous CNS disease in a clinically relevant, humanized, T cell receptor transgenic mouse model. Mice develop spontaneous, ascending paralysis, allowing unbiased characterization of T cell immunity in an HLA-DR15-restricted T cell repertoire. Analysis of naturally progressing disease shows that IFNγ(+) cells dominate disease initiation with IL-17(+) cells apparent in affected tissue only once disease is established. Tregs accumulate in the CNS but are ultimately ineffective at halting disease progression. However, ablation of Tregs causes profound acceleration of disease, with uncontrolled infiltration of lymphocytes into the CNS. This synchronous, severe disease allows characterization of the responses that are deregulated in exacerbated disease: the correlation is with increased CNS CD4 and CD8 IFNγ responses. Recovery of the ablated Treg population halts ongoing disease progression and Tregs extracted from the central nervous system at peak disease are functionally competent to regulate myelin specific T cell responses. Thus, in a clinically relevant mouse model of MS, initial disease is IFNγ driven and the enhanced central nervous system responses unleashed through Treg ablation comprise IFNγ cytokine production by CD4 and CD8 cells, but not IL-17 responses.


Subject(s)
Multiple Sclerosis/pathology , T-Lymphocytes, Regulatory/physiology , Th1 Cells/physiology , Th17 Cells/physiology , Animals , Brain/pathology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Proliferation , Central Nervous System/pathology , Disease Progression , Fluorescent Antibody Technique , HLA-DR Serological Subtypes/metabolism , Humans , Interferon-gamma/physiology , Interleukin-17/metabolism , Mice , Mice, Transgenic , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Real-Time Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics
15.
Pathogens ; 12(8)2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37624013

ABSTRACT

Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen and the leading cause of infection in patients with cystic fibrosis (CF). The ability of P. aeruginosa to evade host responses and develop into chronic infection causes significant morbidity and mortality. Several mouse models have been developed to study chronic respiratory infections induced by P. aeruginosa, with the bead agar model being the most widely used. However, this model has several limitations, including the requirement for surgical procedures and high mortality rates. Herein, we describe novel and adapted biologically relevant models of chronic lung infection caused by P. aeruginosa. Three methods are described: a clinical isolate infection model, utilising isolates obtained from patients with CF; an incomplete antibiotic clearance model, leading to bacterial bounce-back; and the establishment of chronic infection; and an adapted water bottle chronic infection model. These models circumvent the requirement for a surgical procedure and, importantly, can be induced with clinical isolates of P. aeruginosa and in wild-type mice. We also demonstrate successful induction of chronic infection in the transgenic ßENaC murine model of CF. We envisage that the models described will facilitate the investigations of host and microbial factors, and the efficacy of novel antimicrobials, during chronic P. aeruginosa respiratory infections.

16.
Cell Rep ; 42(8): 113012, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37598340

ABSTRACT

How the opportunistic Gram-negative pathogens of the genus Achromobacter interact with the innate immune system is poorly understood. Using three Achromobacter clinical isolates from two species, we show that the type 3 secretion system (T3SS) is required to induce cell death in human macrophages by inflammasome-dependent pyroptosis. Macrophages deficient in the inflammasome sensors NLRC4 or NLRP3 undergo pyroptosis upon bacterial internalization, but those deficient in both NLRC4 and NLRP3 do not, suggesting either sensor mediates pyroptosis in a T3SS-dependent manner. Detailed analysis of the intracellular trafficking of one isolate indicates that the intracellular bacteria reside in a late phagolysosome. Using an intranasal mouse infection model, we observe that Achromobacter damages lung structure and causes severe illness, contingent on a functional T3SS. Together, we demonstrate that Achromobacter species can survive phagocytosis by promoting macrophage cell death and inflammation by redundant mechanisms of pyroptosis induction in a T3SS-dependent manner.


Subject(s)
Achromobacter , Pyroptosis , Humans , Animals , Mice , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Type III Secretion Systems , Disease Models, Animal , Calcium-Binding Proteins , CARD Signaling Adaptor Proteins
17.
Immunology ; 136(3): 325-33, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22384827

ABSTRACT

Streptococcus pyogenes is the causative agent of numerous diseases ranging from benign infections (pharyngitis and impetigo) to severe infections associated with high mortality (necrotizing fasciitis and bacterial sepsis). As with other bacterial infections, there is considerable interest in characterizing the contribution of interleukin-17A (IL-17A) responses to protective immunity. We here show significant il17a up-regulation by quantitative real-time PCR in secondary lymphoid organs, correlating with increased protein levels in the serum within a short time of S. pyogenes infection. However, our data offer an important caveat to studies of IL-17A responsiveness following antigen inoculation, because enhanced levels of IL-17A were also detected in the serum of sham-infected mice, indicating that inoculation trauma alone can stimulate the production of this cytokine. This highlights the potency and speed of innate IL-17A immune responses after inoculation and the importance of proper and appropriate controls in comparative analysis of immune responses observed during microbial infection.


Subject(s)
Interleukin-17/blood , Interleukin-17/genetics , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Adaptive Immunity/genetics , Animals , Antigens, Bacterial/administration & dosage , Base Sequence , DNA Primers/genetics , Female , HLA-DQ Antigens/genetics , Humans , Immunity, Innate/genetics , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Sepsis/genetics , Sepsis/immunology , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Th17 Cells/immunology , Time Factors , Up-Regulation
18.
J Immunol ; 184(7): 3814-21, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20208010

ABSTRACT

There has been a long history of defining T cell epitopes to track viral immunity and to design rational vaccines, yet few data of this type exist for bacterial infections. Bacillus anthracis, the causative agent of anthrax, is both an endemic pathogen in many regions and a potential biological warfare threat. T cell immunity in naturally infected anthrax patients has not previously been characterized, which is surprising given concern about the ability of anthrax toxins to subvert or ablate adaptive immunity. We investigated CD4 T cell responses in patients from the Kayseri region of Turkey who were previously infected with cutaneous anthrax. Responses to B. anthracis protective Ag and lethal factor (LF) were investigated at the protein, domain, and epitope level. Several years after antibiotic-treated anthrax infection, strong T cell memory was detectable, with no evidence of the expected impairment in specific immunity. Although serological responses to existing anthrax vaccines focus primarily on protective Ag, the major target of T cell immunity in infected individuals and anthrax-vaccinated donors was LF, notably domain IV. Some of these anthrax epitopes showed broad binding to several HLA class alleles, but others were more constrained in their HLA binding patterns. Of specific CD4 T cell epitopes targeted within LF domain IV, one is preferentially seen in the context of bacterial infection, as opposed to vaccination, suggesting that studies of this type will be important in understanding how the human immune system confronts serious bacterial infection.


Subject(s)
Anthrax/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Anthrax Vaccines/immunology , Bacillus anthracis/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunologic Memory
19.
Cell Rep ; 40(6): 111167, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35947948

ABSTRACT

Many bacterial pathogens antagonize host defense responses by translocating effector proteins into cells. It remains an open question how those pathogens not encoding effectors counteract anti-bacterial immunity. Here, we show that Klebsiella pneumoniae exploits the evolutionary conserved innate protein SARM1 to regulate negatively MyD88- and TRIF-governed inflammation, and the activation of the MAP kinases ERK and JNK. SARM1 is required for Klebsiella induction of interleukin-10 (IL-10) by fine-tuning the p38-type I interferon (IFN) axis. SARM1 inhibits the activation of Klebsiella-induced absent in melanoma 2 inflammasome to limit IL-1ß production, suppressing further inflammation. Klebsiella exploits type I IFNs to induce SARM1 in a capsule and lipopolysaccharide O-polysaccharide-dependent manner via the TLR4-TRAM-TRIF-IRF3-IFNAR1 pathway. Absence of SARM1 reduces the intracellular survival of K. pneumoniae in macrophages, whereas sarm1-deficient mice control the infection. Altogether, our results illustrate an anti-immunology strategy deployed by a human pathogen. SARM1 inhibition will show a beneficial effect to treat Klebsiella infections.


Subject(s)
Klebsiella Infections , Klebsiella pneumoniae , Adaptor Proteins, Vesicular Transport , Animals , Armadillo Domain Proteins/genetics , Cytoskeletal Proteins , Humans , Inflammation , Mice , Signal Transduction
20.
Biomolecules ; 12(12)2022 11 22.
Article in English | MEDLINE | ID: mdl-36551159

ABSTRACT

Secretory leucoprotease inhibitor (SLPI) has multifaceted functions, including inhibition of protease activity, antimicrobial functions, and anti-inflammatory properties. In this study, we show that SLPI plays a role in controlling pulmonary Pseudomonas aeruginosa infection. Mice lacking SLPI were highly susceptible to P. aeruginosa infection, however there was no difference in bacterial burden. Utilising a model of P. aeruginosa LPS-induced lung inflammation, human recombinant SLPI (hrSLPI) administered intraperitoneally suppressed the recruitment of inflammatory cells in the bronchoalveolar lavage fluid (BALF) and resulted in reduced BALF and serum levels of inflammatory cytokines and chemokines. This anti-inflammatory effect of hrSLPI was similarly demonstrated in a systemic inflammation model induced by intraperitoneal injection of LPS from various bacteria or lipoteichoic acid, highlighting the broad anti-inflammatory properties of hrSLPI. Moreover, in bone-marrow-derived macrophages, hrSLPI reduced LPS-induced phosphorylation of p-IkB-α, p-IKK-α/ß, p-P38, demonstrating that the anti-inflammatory effect of hrSLPI was due to the inhibition of the NFκB and MAPK pathways. In conclusion, administration of hrSLPI attenuates excessive inflammatory responses and is therefore, a promising strategy to target inflammatory diseases such as acute respiratory distress syndrome or sepsis and could potentially be used to augment antibiotic treatment.


Subject(s)
Inflammation , Pseudomonas Infections , Secretory Leukocyte Peptidase Inhibitor , Animals , Humans , Mice , Inflammation/metabolism , Inflammation/microbiology , Lipopolysaccharides , Pseudomonas Infections/metabolism , Pseudomonas Infections/therapy , Secretory Leukocyte Peptidase Inhibitor/administration & dosage , Secretory Leukocyte Peptidase Inhibitor/metabolism , Recombinant Proteins/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL