32 research outputs found

    Can combined cellular and 11beta hydroxysteroid dehydrogenase type 1 gene therapy attenuate inflammation in Acute Respiratory Distress Syndrome?

    Get PDF
    Acute Respiratory Distress Syndrome (ARDS) is an inflammatory disorder of the lungs, for which there is no effective pharmacotherapy. 11β hydroxysteroid dehydrogenase type 1 (HSD-1) acts as a reductase to convert cortisone into cortisol, and is normally expressed by alveolar macrophages (AMs). A previous study has shown evidence of impaired alveolar HSD-1 reductase activity and relative alveolar cortisol deficiency in ARDS patients. Mesenchymal stem cells (MSCs) administration has been effective at attenuating inflammation in pre-clinical models of lung injury. This thesis reports a series of investigations into the role of AM dysfunction in ARDS pathogenesis, and whether use of transgenic MSCs (tMSCs) expressing HSD-1 offers a potential therapy for ARDS by enhancing AM efferocytosis. It also investigates the impact of expansion on the ability of MSCs to attenuate lung injury. Expansion of MSCs for 1 month had no effect on the ability of MSCs to promote alveolar epithelial wound healing, however the ability of MSCs to stimulate IL-10 release from macrophages was impaired. Lentiviral transfection was used to create HSD-1 tMSCs, which expressed functional HSD-1. Development of sepsis-related ARDS is associated with decreased AM HSD-1 reductase activity and efferocytosis, which contributes to the increased alveolar neutrophil apoptosis, alveolar inflammation and mortality observed. Dysregulated AM function contributes to ARDS pathogenesis, therefore upregulation of AM efferocytosis may offer a therapeutic strategy for ARDS patients. ARDS broncho-alveolar lavage fluid (BALF) treatment of healthy AMs in vitro can decrease rac1 gene expression and impair efferocytosis, thereby replicating the AM functional defect observed in ARDS patients. Following ARDS BALF treatment, AMs increase expression of pro-efferocytosis receptors CD206 and MerTK, as an unsuccessful compensatory mechanism to restore normal efferocytosis. HSD-1 tMSC co-culture was unsuccessful at restoring AM efferocytosis both ex vivo and in an in vitro model of ARDS, indicating that HSD-1 independent mechanisms contribute to impaired AM efferocytosis in ARDS. HSD-1 tMSC therapy reduced cellular inflammation in a model of murine peritonitis, but also caused bacterial overgrowth due to immunosuppression, indicating that HSD-1 tMSC therapy is unlikely to have utility in patients with sepsis-related ARDS. However, further studies using multiple low doses of HSD-1 tMSCs in HSD-1 knockout mice in conjunction with antibiotic therapy are required to fully assess the therapeutic potential of HSD-1 tMSCs. Rho-associated protein kinase (ROCK) inhibitor treatment partially restored AM efferocytosis in an in vitro model of ARDS, indicating that targeting of the ROCK pathway may offer therapeutic potential in ARDS patients

    Raised FGF23 Correlates to Increased Mortality in Critical Illness, Independent of Vitamin D

    Get PDF
    BACKGROUND: Fibroblast Growth Factor (FGF23) is an endocrine hormone classically associated with the homeostasis of vitamin D, phosphate, and calcium. Elevated serum FGF23 is a known independent risk factor for mortality in chronic kidney disease (CKD) patients. We aimed to determine if there was a similar relationship between FGF23 levels and mortality in critically ill patients.METHODS: Plasma FGF23 levels were measured by ELISA in two separate cohorts of patients receiving vitamin D supplementation: critical illness patients (VITdAL-ICU trial, n = 475) and elective oesophagectomy patients (VINDALOO trial, n = 76). Mortality data were recorded at 30 and 180 days or at two years, respectively. FGF23 levels in a healthy control cohort were also measured ( n = 27). RESULTS: Elevated FGF23 (quartile 4 vs. quartiles 1-3) was associated with increased short-term (30 and 180 day) mortality in critical illness patients ( p &lt; 0.001) and long-term (two-year) mortality in oesophagectomy patients ( p = 0.0149). Patients who died had significantly higher FGF23 levels than those who survived: In the critical illness cohort, those who died had 1194.6 pg/mL (range 0-14,000), while those who survived had 120.4 pg/mL (range = 15-14,000) ( p = 0.0462). In the oesophagectomy cohort, those who died had 1304 pg/mL (range = 154-77,800), while those who survived had 644 pg/mL (range = 179-54,894) ( p &lt; 0.001). This was found to be independent of vitamin D or CKD status (critical illness p = 0.3507; oesophagectomy p = 0.3800). FGF23 levels in healthy controls were similar to those seen in oesophagectomy patients ( p = 0.4802). CONCLUSIONS: Elevated baseline serum FGF23 is correlated with increased mortality in both the post-oesophagectomy cohort and the cohort of patients with critical illness requiring intensive care admission. This was independent of vitamin D status, supplementation, or CKD status, which suggests the presence of vitamin D-independent mechanisms of FGF23 action during the acute and convalescent stages of critical illness, warranting further investigation.</p

    Nocardia kroppenstedtii sp. nov., a novel actinomycete isolated from a lung transplant patient with a pulmonary infection

    Get PDF
    An actinomycete, strain N1286T, isolated from a lung transplant patient with a pulmonary infection, was provisionally assigned to the genus Nocardia. The strain had chemotaxonomic and morphological properties typical of members of the genus Nocardia and formed a distinct phyletic line in the Nocardia 16S rRNA gene tree. It was most closely related to Nocardia farcinica DSM 43665T (99.8% gene similarity) but was distinguished from the latter by a low level of DNA:DNA relatedness. These strains were also distinguished by a broad range of phenotypic properties. On the basis of these data, it is proposed that isolate N1286T (=DSM 45810T = NCTC 13617T) should be classified as the type strain of a new Nocardia species for which the name Nocardia kroppenstedtii is proposed

    Simvastatin to modify neutrophil function in older patients with septic pneumonia (SNOOPI) : study protocol for a randomised placebo-controlled trial

    Get PDF
    BACKGROUND: Community-acquired pneumonia (CAP) is considered the leading cause of death from infectious disease in developed countries, while complications of CAP - sepsis being the most common and challenging - increase the risk of mortality. During the progression of sepsis, a state of neutrophil ‘paralysis’ develops resulting in the impairment of neutrophil anti-microbial functions including: chemotaxis, production of reactive oxygen species, and formation of neutrophil extracellular traps (NETs). Mechanisms underlying defective neutrophil function remain elusive although NET formation has been implicated in the immunosuppression and increased rates of sepsis observed in neonates. There is, however, increasing evidence that statins are able to modulate neutrophil function in sepsis as several systematic reviews have concluded that statins have a role in improving infection-related outcomes and mortality while, in vitro, statins have also been shown to boost NET formation in healthy individuals. METHODS/DESIGN: The ‘SNOOPI’ trial is a phase 4, randomised placebo-controlled trial. The aim of this study is to determine whether oral treatment with simvastatin compared to placebo optimises neutrophil anti-microbial functions in elderly patients with septic pneumonia improving patient outcomes in the elderly. The primary outcome will be NET production within 72 to 96 hours of treatment with simvastatin or placebo measured in response to a number of inflammatory mediators, including IL8, f-Met-Leu-Phe and lipopolysaccharide. Secondary outcomes include neutrophil migratory capacity; reactive oxygen species production; neutrophil phagocytic capacity; safety and tolerability of simvastatin administration within this patient group; biological markers of neutrophil activation, the inflammatory response, alveolar epithelial and endothelial injury; systemic endothelial function biomarkers and pulmonary extracellular matrix degradation. This study aims to recruit 60 patients admitted into Queen Elizabeth Hospital Birmingham NHS-Foundation Trust. DISCUSSION: This study will investigate the ability of in vivo simvastatin therapy to modulate neutrophil anti-microbial functions in CAP-associated sepsis. TRIAL REGISTRATION: EudraCT number: 2012-003343-29 (Trial Registered: 26 November 2012). ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/1745-6215-15-332) contains supplementary material, which is available to authorized users

    Dexamethasone impairs the expression of antimicrobial mediators in lipopolysaccharide-activated primary macrophages by inhibiting both expression and function of interferon β

    Get PDF
    Glucocorticoids potently inhibit expression of many inflammatory mediators, and have been widely used to treat both acute and chronic inflammatory diseases for more than seventy years. However, they can have several unwanted effects, amongst which immunosuppression is one of the most common. Here we used microarrays and proteomic approaches to characterise the effect of dexamethasone (a synthetic glucocorticoid) on the responses of primary mouse macrophages to a potent pro-inflammatory agonist, lipopolysaccharide (LPS). Gene ontology analysis revealed that dexamethasone strongly impaired the lipopolysaccharide-induced antimicrobial response, which is thought to be driven by an autocrine feedback loop involving the type I interferon IFNβ. Indeed, dexamethasone strongly and dose-dependently inhibited the expression of IFNβ by LPS-activated macrophages. Unbiased proteomic data also revealed an inhibitory effect of dexamethasone on the IFNβ-dependent program of gene expression, with strong down-regulation of several interferon-induced antimicrobial factors. Surprisingly, dexamethasone also inhibited the expression of several antimicrobial genes in response to direct stimulation of macrophages with IFNβ. We tested a number of hypotheses based on previous publications, but found that no single mechanism could account for more than a small fraction of the broad suppressive impact of dexamethasone on macrophage type I interferon signaling, underlining the complexity of this pathway. Preliminary experiments indicated that dexamethasone exerted similar inhibitory effects on primary human monocyte-derived or alveolar macrophages.</p

    Vitamin D to prevent lung injury following esophagectomy: A randomized, placebo-controlled trial

    Get PDF
    Objectives: Observational studies suggest an association between vitamin D deficiency and adverse outcomes of critical illness and identify it as a potential risk factor for the development of lung injury. To determine whether pre-operative administration of oral high-dose cholecalciferol ameliorates early acute lung injury post-operatively in adults undergoing elective esophagectomy. Design: A double-blind, randomized, placebo-controlled trial. Setting: Three large UK university hospitals. Patients: Seventy-nine adult patients undergoing elective esophagectomy were randomized. Intervention: A single oral preoperative (3-14 days) dose of 7.5mg (300,000IU; 15mls) cholecalciferol or matched placebo. Measurements and Main Results: Primary outcome was change in extravascular lung water index (EVLWI) at the end of esophagectomy. Secondary outcomes included PaO2:FiO2 ratio, development of lung injury, ventilator and organ-failure free days, 28 and 90 day survival, safety of cholecalciferol supplementation, plasma vitamin D status (25(OH)D, 1,25(OH)2D and vitamin D binding protein), pulmonary vascular permeability index (PVPI) and EVLWI day 1 postoperatively. An exploratory study measured biomarkers of alveolar-capillary inflammation and injury. Forty patients were randomized to cholecalciferol and 39 to placebo. There was no significant change in EVLWI at the end of the operation between treatment groups (placebo median 1.0[IQR 0.4 – 1.8] vs cholecalciferol median 0.4[IQR 0.4 – 1.2] ml/kg, p=0.059). Median PVPI values were significantly lower in the cholecalciferol treatment group (placebo 0.4[IQR 0 – 0.7] vs cholecalciferol 0.1[IQR -0.15 -0.35], p=0.027). Cholecalciferol treatment effectively increased 25(OH)D concentrations but surgery resulted in a decrease in 25(OH)D concentrations at day 3 in both arms. There was no difference in clinical outcomes. Conclusions: High-dose preoperative treatment with oral cholecalciferol was effective at increasing 25(OH)D concentrations, and reduced changes in postoperative PVPI but not EVLWI
    corecore