Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
Front Pharmacol ; 13: 887608, 2022.
Article in English | MEDLINE | ID: mdl-35814225

ABSTRACT

Chemotherapy causes sensory disturbances in cancer patients that results in neuropathies and pain. As cancer survivorships has dramatically increased over the past 10 years, pain management of these patients is becoming clinically more important. Current analgesic strategies are mainly ineffective and long-term use is associated with severe side effects. The issue being that common analgesic strategies are based on ubiquitous pain mediator pathways, so when applied to clinically diverse neuropathic pain and neurological conditions, are unsuccessful. This is principally due to the lack of understanding of the driving forces that lead to chemotherapy induced neuropathies. It is well documented that chemotherapy causes sensory neurodegeneration through axonal atrophy and intraepidermal fibre degeneration causing alterations in pain perception. Despite the neuropathological alterations associated with chemotherapy-induced neuropathic pain being extensively researched, underlying causes remain elusive. Resent evidence from patient and rodent studies have indicated a prominent inflammatory cell component in the peripheral sensory nervous system in effected areas post chemotherapeutic treatment. This is accompanied by modulation of auxiliary cells of the dorsal root ganglia sensory neurons such as activation of satellite glia and capillary dysfunction. The presence of a neuroinflammatory component was supported by transcriptomic analysis of dorsal root ganglia taken from mice treated with common chemotherapy agents. With key inflammatory mediators identified, having potent immunoregulatory effects that directly influences nociception. We aim to evaluate the current understanding of these immune-neuronal interactions across different cancer therapy drug classes. In the belief this may lead to better pain management approaches for cancer survivors.

3.
Sci Rep ; 9(1): 9218, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31239465

ABSTRACT

A low quadriceps slow-twitch (ST), oxidative (relative to fast-twitch) fiber proportion is prevalent in chronic diseases such Chronic Obstructive Pulmonary Disease (COPD) and is associated with exercise limitation and poor outcomes. Benefits of an increased ST fiber proportion are demonstrated in genetically modified animals. Pathway analysis of published data of differentially expressed genes in mouse ST and FT fibers, mining of our microarray data and a qPCR analysis of quadriceps specimens from COPD patients and controls were performed. ST markers were quantified in C2C12 myotubes with EGF-neutralizing antibody, EGFR inhibitor or an EGFR-silencing RNA added. A zebrafish egfra mutant was generated by genome editing and ST fibers counted. EGF signaling was (negatively) associated with the ST muscle phenotype in mice and humans, and muscle EGF transcript levels were raised in COPD. In C2C12 myotubes, EGFR inhibition/silencing increased ST, including mitochondrial, markers. In zebrafish, egfra depletion increased ST fibers and mitochondrial content. EGF is negatively associated with ST muscle phenotype in mice, healthy humans and COPD patients. EGFR blockade promotes the ST phenotype in myotubes and zebrafish embryos. EGF signaling suppresses the ST phenotype, therefore EGFR inhibitors may be potential treatments for COPD-related muscle ST fiber loss.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Aged , Animals , Case-Control Studies , Epidermal Growth Factor/genetics , Female , Humans , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Middle Aged , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Oxidation-Reduction/drug effects , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , RNA, Messenger/genetics , Zebrafish
4.
Sci Rep ; 8(1): 13987, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228360

ABSTRACT

Amine quantification is an important strategy in patient stratification and personalised medicine. This is because amines, including amino acids and methylarginines impact on many homeostatic processes. One important pathway regulated by amine levels is nitric oxide synthase (NOS). NOS is regulated by levels of (i) the substrate, arginine, (ii) amino acids which cycle with arginine and (iii) methylarginine inhibitors of NOS. However, biomarker research in this area is hindered by the lack of a unified analytical platform. Thus, the development of a common metabolomics platform, where a wide range of amino acids and methylarginines can be measured constitutes an important unmet need. Here we report a novel high-throughput ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) platform where ≈40 amine analytes, including arginine and methylarginines can be detected and quantified on a molar basis, in a single sample of human plasma. To validate the platform and to generate biomarkers, human plasma from a well-defined cohort of patients before and after coronary artery bypass surgery, who developed systemic inflammatory response syndrome (SIRS), were analysed. Bypass surgery with SIRS significantly altered 26 amine analytes, including arginine and ADMA. Consequently, pathway analysis revealed significant changes in a range of pathways including those associated with NOS.


Subject(s)
Amines/blood , Amino Acids/blood , Arginine/analogs & derivatives , Biomarkers/blood , Chromatography, High Pressure Liquid/methods , Systemic Inflammatory Response Syndrome/blood , Tandem Mass Spectrometry/methods , Aged , Arginine/blood , Female , Humans , Male , Prognosis , Systemic Inflammatory Response Syndrome/surgery
5.
PLoS One ; 12(8): e0183025, 2017.
Article in English | MEDLINE | ID: mdl-28813479

ABSTRACT

RATIONALE: Nitric oxide synthase (NOS) is a biomarker/target in sepsis. NOS activity is driven by amino acids, which cycle to regulate the substrate L-arginine in parallel with cycles which regulate the endogenous inhibitors ADMA and L-NMMA. The relationship between amines and the consequence of plasma changes on iNOS activity in early sepsis is not known. OBJECTIVE: Our objective was to apply a metabolomics approach to determine the influence of sepsis on a full array of amines and what consequence these changes may have on predicted iNOS activity. METHODS AND MEASUREMENTS: 34 amino acids were measured using ultra purification mass spectrometry in the plasma of septic patients (n = 38) taken at the time of diagnosis and 24-72 hours post diagnosis and of healthy volunteers (n = 21). L-arginine and methylarginines were measured using liquid-chromatography mass spectrometry and ELISA. A top down approach was also taken to examine the most changed metabolic pathways by Ingenuity Pathway Analysis. The iNOS supporting capacity of plasma was determined using a mouse macrophage cell-based bioassay. MAIN RESULTS: Of all the amines measured 22, including L-arginine and ADMA, displayed significant differences in samples from patients with sepsis. The functional consequence of increased ADMA and decreased L-arginine in context of all cumulative metabolic changes in plasma resulted in reduced iNOS supporting activity associated with sepsis. CONCLUSIONS: In early sepsis profound changes in amine levels were defined by dominant changes in the iNOS canonical pathway resulting in functionally meaningful changes in the ability of plasma to regulate iNOS activity ex vivo.


Subject(s)
Amines/metabolism , Metabolomics , Sepsis/metabolism , Adult , Aged , Animals , Arginine/metabolism , Cell Line , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mass Spectrometry , Mice , Middle Aged , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/metabolism , Sepsis/physiopathology , omega-N-Methylarginine/metabolism
6.
FASEB J ; 31(6): 2439-2445, 2017 06.
Article in English | MEDLINE | ID: mdl-28246166

ABSTRACT

IL-1ß release is integral to the innate immune system. The release of mature IL-1ß depends on 2 regulated events: the de novo induction of pro-IL-1ß, generally via NF-κB-dependent transduction pathways; and the assembly and activation of the NLRP3 inflammasome. This latter step is reliant on active caspase-1, pannexin-1, and P2X7 receptor activation. Pathogen-associated molecular patterns in gram-positive and gram-negative bacteria activate IL-1ß release from immune cells via TLR2 and TLR4 receptors, respectively. We found that pro-IL-1ß and mature IL-1ß release from human monocytes is stimulated by the TLR2 agonists Pam3CSK4 or FSL-1, as well as the TLR4 agonist LPS in the absence of additional ATP. TLR2 agonists required pannexin-1 and P2X7 receptor activation to stimulate IL-1ß release. In contrast, IL-1ß release stimulated by the TLR4 agonist LPS is independent of both pannexin-1 and P2X7 activation. In the absence of exogenous ATP, P2X7 activation requires endogenous ATP release, which occurs in some cells via pannexin-1. In line with this, we found that LPS-stimulated human monocytes released relatively low levels of ATP, whereas cells stimulated with TLR2 agonists released high levels of ATP. These findings suggest that in human monocytes, both TLR2 and TLR4 signaling induce pro-IL-1ß expression, but the mechanism by which they activate caspase-1 diverges at the level of the pannexin-1/ATP/P2X7 axis.-Parzych, K., Zetterqvist, A. V., Wright, W. R., Kirkby, N. S., Mitchell, J. A., Paul-Clark, M. J. Differential role of pannexin-1/ATP/P2X7 axis in IL-1ß release by human monocytes.


Subject(s)
Adenosine Triphosphate/metabolism , Connexins/metabolism , Interleukin-1beta/metabolism , Monocytes/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Adenosine Triphosphate/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cell Line, Tumor , Connexins/genetics , Diglycerides/pharmacology , Gene Expression Regulation/physiology , Humans , Interleukin-1beta/genetics , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Nerve Tissue Proteins/genetics , Oligopeptides/pharmacology , Receptors, Purinergic P2X7/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
7.
PLoS One ; 11(2): e0147289, 2016.
Article in English | MEDLINE | ID: mdl-26910063

ABSTRACT

Nonsteroidal anti-inflammatory drugs have been shown to reduce the incidence of gastrointestinal cancers, but the propensity of these drugs to cause ulcers and bleeding limits their use. H2S has been shown to be a powerful cytoprotective and anti-inflammatory substance in the digestive system. This study explored the possibility that a H2S-releasing nonsteroidal anti-inflammatory drug (ATB-346) would be effective in a murine model of hereditary intestinal cancer (APCMin+ mouse) and investigated potential mechanisms of action via transcriptomics analysis. Daily treatment with ATB-346 was significantly more effective at preventing intestinal polyp formation than naproxen. Significant beneficial effects were seen with a treatment period of only 3-7 days, and reversal of existing polyps was observed in the colon. ATB-346, but not naproxen, significantly decreased expression of intestinal cancer-associated signaling molecules (cMyc, ß-catenin). Transcriptomic analysis identified 20 genes that were up-regulated in APCMin+ mice, 18 of which were reduced to wild-type levels by one week of treatment with ATB-346. ATB-346 is a novel, gastrointestinal-sparing anti-inflammatory drug that potently and rapidly prevents and reverses the development of pre-cancerous lesions in a mouse model of hereditary intestinal tumorigenesis. These effects may be related to the combined effects of suppression of cyclooxygenase and release of H2S, and correction of most of the APCMin+-associated alterations in the transcriptome. ATB-346 may represent a promising agent for chemoprevention of tumorigenesis in the GI tract and elsewhere.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Carcinogenesis/drug effects , Hydrogen Sulfide/chemistry , Intestinal Neoplasms/pathology , Intestinal Neoplasms/prevention & control , Naproxen/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chemoprevention , Colon/drug effects , Colon/metabolism , Colon/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Intestinal Polyps/prevention & control , Male , Mice , Mice, Inbred C57BL , Naproxen/chemistry , Naproxen/therapeutic use , Proto-Oncogene Proteins c-myc/metabolism , beta Catenin/metabolism
8.
BMJ Open ; 5(1): e005750, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25631307

ABSTRACT

INTRODUCTION: Cigarette smoke contributes to a diverse range of diseases including chronic obstructive pulmonary disease (COPD), cardiovascular disorders and many cancers. There currently is a need for human challenge models, to assess the acute effects of a controlled cigarette smoke stimulus, followed by serial sampling of blood and respiratory tissue for advanced molecular profiling. We employ precision sampling of nasal mucosal lining fluid by absorption to permit soluble mediators measurement in eluates. Serial nasal curettage was used for transcriptomic analysis of mucosal tissue. METHODS AND ANALYSIS: Three groups of strictly defined patients will be studied: 12 smokers with COPD (GOLD Stage 2) with emphysema, 12 matched smokers with normal lung function and no evidence of emphysema, and 12 matched never smokers with normal spirometry. Patients in the smoking groups are current smokers, and will be given full support to stop smoking immediately after this study. In giving a controlled cigarette smoke stimulus, all patients will have abstained from smoking for 12 h, and will smoke two cigarettes with expiration through the nose in a ventilated chamber. Before and after inhalation of cigarette smoke, a series of samples will be taken from the blood, nasal mucosal lining fluid and nasal tissue by curettage. Analysis of plasma nicotine and metabolites in relation to levels of soluble inflammatory mediators in nasal lining fluid and blood, as well as assessing nasal transcriptomics, ex vivo blood platelet aggregation and leucocyte responses to toll-like receptor agonists will be undertaken. IMPLICATIONS: Development of acute cigarette smoke challenge models has promise for the study of molecular effects of smoking in a range of pathological processes. ETHICS AND DISSEMINATION: This study was approved by the West London National Research Ethics Committee (12/LO/1101). The study findings will be presented at conferences and will be reported in peer-reviewed journals.


Subject(s)
Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Research Design , Smoking/immunology , Smoking/metabolism , Administration, Inhalation , Humans , Models, Biological , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Pulmonary Emphysema/immunology , Pulmonary Emphysema/metabolism
9.
PLoS One ; 9(6): e98165, 2014.
Article in English | MEDLINE | ID: mdl-24887395

ABSTRACT

Cyxlo-oxygenase (COX)-2 inhibitors, including traditional nonsteroidal anti-inflammatory drugs (NSAIDs) are associated with increased cardiovascular side effects, including myocardial infarction. We and others have shown that COX-1 and not COX-2 drives vascular prostacyclin in the healthy cardiovascular system, re-opening the question of how COX-2 might regulate cardiovascular health. In diseased, atherosclerotic vessels, the relative contribution of COX-2 to prostacyclin formation is not clear. Here we have used apoE(-/-)/COX-2(-/-) mice to show that, whilst COX-2 profoundly limits atherosclerosis, this protection is independent of local prostacyclin release. These data further illustrate the need to look for new explanations, targets and pathways to define the COX/NSAID/cardiovascular risk axis. Gene expression profiles in tissues from apoE(-/-)/COX-2(-/-) mice showed increased lymphocyte pathways that were validated by showing increased T-lymphocytes in plaques and elevated plasma Th1-type cytokines. In addition, we identified a novel target gene, rgl1, whose expression was strongly reduced by COX-2 deletion across all examined tissues. This study is the first to demonstrate that COX-2 protects vessels against atherosclerotic lesions independently of local vascular prostacyclin and uses systems biology approaches to identify new mechanisms relevant to development of next generation NSAIDs.


Subject(s)
Atherosclerosis/enzymology , Blood Vessels/metabolism , Cyclooxygenase 2/metabolism , Epoprostenol/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protective Agents/metabolism , T-Lymphocytes/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Blood Vessels/pathology , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Female , Gene Deletion , Gene Expression Regulation, Enzymologic , Male , Mice, Inbred C57BL , Signal Transduction , Transcriptome/genetics , Up-Regulation/genetics
10.
Article in English | MEDLINE | ID: mdl-23742950

ABSTRACT

Cyclooxygenase (COX) is required for prostanoid (e.g. prostaglandin PGE2) production. Constitutive COX-1 and inducible COX-2 are implicated in lung diseases, such as idiopathic pulmonary fibrosis (IPF). Using lung fibroblasts from humans and wild type, COX-1(-/-) and COX-2(-/-) mice, we investigated how COX activity modulates cell growth and inflammatory responses induced by activators of Toll-like receptors (TLRs) 1-8. In mouse tissue, PGE2 release from fresh lung was COX-1 driven, in lung in culture (24h) COX-1 and COX-2 driven, and from proliferating lung fibroblasts exclusively COX-2 driven. COX-2 limited proliferation in lung fibroblasts and both isoforms limited KC release induced by a range of TLR agonists. Less effect of COX was seen on TLR-induced IP-10 release. In human lung fibroblasts inhibition of COX with diclofenac was associated with increased release of IL-8 and IP-10. Our results may have implications for the treatment of IPF.


Subject(s)
Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytokines/metabolism , Fibroblasts/enzymology , Membrane Proteins/genetics , Toll-Like Receptors/agonists , Animals , Cell Proliferation , Cells, Cultured , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Diclofenac/pharmacology , Dinoprostone/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Host-Pathogen Interactions , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Immunity, Innate , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Toll-Like Receptors/metabolism
11.
Pulm Circ ; 3(4): 926-35, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25006409

ABSTRACT

Pulmonary hypertension is a debilitating disease with no cure. We have previously shown that peroxisome proliferator-activated receptor (PPAR) ß/δ agonists protect the right heart in hypoxia-driven pulmonary hypertension without affecting vascular remodeling. PPARß/δ is an important receptor in lipid metabolism, athletic performance, and the sensing of prostacyclin. Treatment of right heart hypertrophy and failure in pulmonary hypertension is an emerging target for future therapy. Here we have investigated the potential of GW0742, a PPARß agonist, to act directly on the right heart in vivo and what transcriptomic signatures are associated with its actions. Right heart hypertrophy and failure was induced in mice using a pulmonary artery banding (PAB) model. GW0742 was administered throughout the study. Cardiovascular parameters were measured using echocardiography and pressure monitoring. Fibrosis and cellular changes were measured using immunohistochemistry. Transcriptomics were measured using the Illumina MouseRef-8v3 BeadChip array and analyzed using GeneSpring GX (ver. 11.0). PAB resulted in right heart hypertrophy and failure and in increased fibrosis. GW0742 reduced or prevented the effects of PAB on all parameters measured. GW0742 altered a number of genes in the transcriptome, with Angptl4 emerging as the top gene altered (increased) in animals with PAB. In conclusion, the PPARß/δ agonist GW0742 has direct protective effects on the right heart in vivo. These observations identify PPARß/δ as a viable therapeutic target to treat pulmonary hypertension that may complement current and future vasodilator drugs.

12.
Biochem Biophys Res Commun ; 426(4): 486-91, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22960172

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare but fatal condition in which raised pulmonary vascular resistance leads to right heart failure and death. Endothelin-1 is a potent endogenous vasoconstrictor, which is considered to be central to many of the events that lead to PAH, and is an important therapeutic target in the treatment of the condition. In many cases of PAH, the aetiology is unknown but inflammation is increasingly thought to play an important role and viruses have been implicated in the development of disease. The Toll Like Receptors (TLRs) play a key role in innate immune responses by initiating specific anti-bacterial and anti-viral defences in recognition of signature molecular motifs on the surface of invading pathogens. In this study, we set out to examine the expression of bacterial and viral TLRs in human pulmonary artery smooth muscle cells and to establish whether their activation could be relevant to PAH. We found that the viral TLR3 and bacterial TLRs 4 and 6 were most abundantly expressed in human pulmonary artery smooth muscle cells. Using specific TLR ligands, we found that activation of TLRs 3 and 4 resulted in IL-8 release by human pulmonary artery smooth muscle cells but that only TLR3 stimulation resulted in IP10 and endothelin-1 release. These data suggest that human pulmonary artery smooth muscle cells express significant levels of viral TLR3 and respond to its activation by releasing endothelin-1. This may have importance in understanding the association between viruses and the development of PAH.


Subject(s)
Endothelin-1/biosynthesis , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Toll-Like Receptor 3/metabolism , Cells, Cultured , Chemokines/genetics , Cytokines/genetics , Familial Primary Pulmonary Hypertension , Gene Expression , Humans , Hypertension, Pulmonary/virology , Interleukin-8/genetics , Muscle, Smooth, Vascular/virology , Myocytes, Smooth Muscle/virology , Poly I-C/pharmacology , Receptors, Cytokine/genetics , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/genetics , Tumor Necrosis Factor-alpha/pharmacology
14.
PLoS One ; 7(2): e30120, 2012.
Article in English | MEDLINE | ID: mdl-22363418

ABSTRACT

BACKGROUND: Cigarette smoking is responsible for 5 million deaths worldwide each year, and is a major risk factor for cardiovascular and lung diseases. Cigarette smoke contains a complex mixture of over 4000 chemicals containing 10(15) free radicals. Studies show smoke is perceived by cells as an inflammatory and xenobiotic stimulus, which activates an immune response. The specific cellular mechanisms driving cigarette smoke-induced inflammation and disease are not fully understood, although the innate immune system is involved in the pathology of smoking related diseases. METHODOLOGY/PRINCIPLE FINDINGS: To address the impact of smoke as an inflammagen on the innate immune system, THP-1 cells and Human PBMCs were stimulated with 3 and 10% (v/v) cigarette smoke extract (CSE) for 8 and 24 hours. Total RNA was extracted and the transcriptome analysed using Illumina BeadChip arrays. In THP-1 cells, 10% CSE resulted in 80 genes being upregulated and 37 downregulated by ≥1.5 fold after 8 hours. In PBMCs stimulated with 10% CSE for 8 hours, 199 genes were upregulated and 206 genes downregulated by ≥1.5 fold. After 24 hours, the number of genes activated and repressed by ≥1.5 fold had risen to 311 and 306 respectively. The major pathways that were altered are associated with cell survival, such as inducible antioxidants, protein chaperone and folding proteins, and the ubiquitin/proteosome pathway. CONCLUSIONS: Our results suggest that cigarette smoke causes inflammation and has detrimental effects on the metabolism and function of innate immune cells. In addition, THP-1 cells provide a genetically stable alternative to primary cells for the study of the effects of cigarette smoke on human monocytes.


Subject(s)
Gene Expression Profiling/methods , Inflammation/genetics , Monocytes/metabolism , Monocytes/pathology , Smoking/genetics , Cell Line , Databases as Topic , Gene Expression Regulation , Gene Regulatory Networks/genetics , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Principal Component Analysis , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Am J Respir Crit Care Med ; 184(11): 1261-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21868501

ABSTRACT

RATIONALE: Children with congenital heart disease are at risk of gut barrier dysfunction and translocation of gut bacterial antigens into the bloodstream. This may contribute to inflammatory activation and organ dysfunction postoperatively. OBJECTIVES: To investigate the role of intestinal injury and endotoxemia in the pathogenesis of organ dysfunction after surgery for congenital heart disease. METHODS: We analyzed blood levels of intestinal fatty acid binding protein and endotoxin (endotoxin activity assay) alongside global transcriptomic profiling and assays of monocyte endotoxin receptor expression in children undergoing surgery for congenital heart disease. MEASUREMENTS AND MAIN RESULTS: Levels of intestinal fatty acid binding protein and endotoxin were greater in children with duct-dependent cardiac lesions. Endotoxemia was associated with severity of vital organ dysfunction and intensive care stay. We identified activation of pathogen-sensing, antigen-processing, and immune-suppressing pathways at the genomic level postoperatively and down-regulation of pathogen-sensing receptors on circulating immune cells. CONCLUSIONS: Children undergoing surgery for congenital heart disease are at increased risk of intestinal mucosal injury and endotoxemia. Endotoxin activity correlates with a number of outcome variables in this population, and may be used to guide the use of gut-protective strategies.


Subject(s)
Endotoxemia/microbiology , Heart Defects, Congenital/surgery , Intestinal Diseases/microbiology , Intestinal Mucosa/injuries , Intestinal Mucosa/microbiology , Down-Regulation/immunology , Endotoxemia/blood , Endotoxemia/immunology , Enzyme-Linked Immunosorbent Assay , Fatty Acid-Binding Proteins/blood , Fatty Acid-Binding Proteins/immunology , Female , Humans , Infant , Inflammation/blood , Inflammation/immunology , Inflammation/microbiology , Intestinal Diseases/blood , Intestinal Diseases/immunology , Intestinal Mucosa/immunology , Length of Stay/statistics & numerical data , Male , Multiple Organ Failure/blood , Multiple Organ Failure/immunology , Multiple Organ Failure/microbiology , Postoperative Complications/blood , Postoperative Complications/immunology , Postoperative Complications/microbiology , Severity of Illness Index
16.
PLoS One ; 5(5): e10501, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20463927

ABSTRACT

Treatment of human disease with human embryonic stem cell (hESC)-derived cells is now close to reality, but little is known of their responses to physiological and pathological insult. The ability of cells to respond via activation of Toll like receptors (TLR) is critical in innate immune sensing in most tissues, but also extends to more general danger sensing, e.g. of oxidative stress, in cardiomyocytes. We used biomarker release and gene-array analysis to compare responses in hESC before and after differentiation, and to those in primary human endothelial cells. The presence of cardiomyocytes and endothelial cells was confirmed in differentiated cultures by immunostaining, FACS-sorting and, for cardiomyocytes, beating activity. Undifferentiated hESC did not respond with CXCL8 release to Gram positive or Gram negative bacteria, or a range of PAMPs (pathogen associated molecular patterns) for TLRs 1-9 (apart from flagellin, an activator of TLR5). Surprisingly, lack of TLR-dependent responses was maintained over 4 months of differentiation of hESC, in cultures which included cardiomyocytes and endothelial cells. In contrast, primary cultures of human aortic endothelial cells (HAEC) demonstrated responses to a broad range of PAMPs. Expression of downstream TLR signalling pathways was demonstrated in hESC, and IL-1beta, TNFalpha and INFgamma, which bypass the TLRs, stimulated CXCL8 release. NFkappaB pathway expression was also present in hESC and NFkappaB was able to translocate to the nucleus. Low expression levels of TLRs were detected in hESC, especially TLRs 1 and 4, explaining the lack of response of hESC to the main TLR signals. TLR5 levels were similar between differentiated hESC and HAEC, and siRNA knockdown of TLR5 abolished the response to flagellin. These findings have potential implications for survival and function of grafted hESC-derived cells.


Subject(s)
Embryonic Stem Cells/immunology , Endothelial Cells/immunology , Immunity, Innate/immunology , Adult , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Escherichia coli/drug effects , Escherichia coli/immunology , Flagellin/immunology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
17.
Arterioscler Thromb Vasc Biol ; 29(5): 706-11, 2009 May.
Article in English | MEDLINE | ID: mdl-19150877

ABSTRACT

OBJECTIVES: Statins and fibrates are hypolipidemic drugs which decrease cardiac events in individuals without raised levels of cholesterol. These drugs inhibit platelet function, but the mechanisms by which this pleiotropic effect is exerted are not known. METHODS AND RESULTS: We used a range of approaches to show statins inhibit human platelet activation in vitro while engaging PPARalpha and PPARgamma. The effects of simvastatin were prevented by the PPARgamma antagonist GW9662 or the PPARalpha antagonist GW6471. In a small-scale human study fluvastatin activated PPARalpha and PPARgamma in platelets and reduced aggregation in response to arachidonic acid ex vivo. The effects of fenofibrate were prevented by PPARalpha antagonism with GW6471. Fenofibrate increased bleeding time in wild-type, but not in PPARalpha-/- mice. The inhibitory effect of fenofibrate, but not simvastatin, on aggregation was prevented by deletion of PPARalpha in murine platelets. PKCalpha, which influences platelet activation, associated and immune-precipitated with PPARgamma in platelets stimulated with statins and with PPARalpha in platelets stimulated with fenofibrate. CONCLUSIONS: This study is the first to provide a unifying explanation of how fibrates and statins reduce thrombotic and cardiovascular risk. Our findings that PPARs associate with PKCalpha in platelets also provide a mechanism by which these effects are mediated.


Subject(s)
Blood Platelets/drug effects , Clofibric Acid/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypolipidemic Agents/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Animals , Blood Platelets/physiology , Humans , Mice , Mice, Knockout , PPAR alpha/drug effects , PPAR gamma/drug effects , Platelet Adhesiveness/drug effects
18.
Am J Respir Crit Care Med ; 179(4): 299-306, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19011150

ABSTRACT

RATIONALE: The mechanisms by which oxidants are sensed by cells and cause inflammation are not well understood. OBJECTIVES: This study aimed to determine how cells "sense" soluble oxidants and how this is translated into an inflammatory reaction. METHODS: Monocytes, macrophages, or HEK293 cells (stably transfected with human Toll-like receptor [TLR]2, TLR2/1, TLR2/6, or TLR4/MD2-CD14) were used. CXC ligand-8 (CXCL8) levels were measured using ELISA. Phosphorylated IL-1 receptor-associated kinase 1 levels were measured using Western blot. TLR2(-/-) and TLR4(-/-) mice were challenged with oxidants, and inflammation was measured by monitoring cell infiltration and KC levels. MEASUREMENTS AND MAIN RESULTS: Oxidants evoked the release of CXCL8 from monocytes/macrophages; this was abrogated by pretreatment with N-acetylcysteine or binding antibodies to TLR2 and was associated with the rapid phosphorylation of IL-1 receptor-associated kinase 1. Oxidants added to HEK293 cells transfected with TLR2, TLR1/2, or TLR2/6 but not TLR4/MD2-CD14 or control HEK nulls resulted in the release of CXCL8. Oxidant challenge delivered intraperitoneally (2-24 hours) or by inhalation to the lungs (3 days) resulted in a robust inflammation in wild-type mice. TLR2(-/-) mice did not respond to oxidant challenge in either model. TLR4(-/-) mice responded as wild-type mice to oxidants at 2 hours but as TLR2(-/-) mice at later time points. CONCLUSIONS: Oxidant-TLR2 interactions provide a signal that initiates the inflammatory response.


Subject(s)
Bronchitis/metabolism , Oxidants/metabolism , Peritonitis/metabolism , Toll-Like Receptor 2/immunology , Animals , Blotting, Western/methods , Bronchitis/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Humans , Mice , Mice, Inbred C57BL , Oxidants/immunology , Oxidative Stress , Peritonitis/immunology , Smoking/immunology , Smoking/metabolism
19.
Mol Med ; 14(5-6): 238-46, 2008.
Article in English | MEDLINE | ID: mdl-18231574

ABSTRACT

We have recently demonstrated that oxidants can activate monocytes via an action on Toll-like receptor (TLR) 2; however, it is unclear what functional consequence this has on immune surveillance for Gram-negative and -positive bacteria. Gram-negative and -positive bacteria and their related pathogen-associated molecular patterns (PAMPs) are sensed by TLR4 and TLR2, respectively. In the current study, we used a human monocyte cell line to show that oxidants prime cells to subsequent challenge with Gram-negative or -positive bacteria as well as PAMPs specific for TLR4 (LPS), TLR2/1 (Pam(3)CSK4), TLR2/6 (FSL-1), Nod1 (FK565), and Nod2 (MDP Lys 18). Similarly, activation of TLR4 with LPS primed for subsequent activation of cells by agonists of the TLR2/6 or TLR2/1 complex. However, no synergy was noted when cells were costimulated with Pam(3)CSK4 and FSL-1. We then tested blood (and isolated monocytes) derived from healthy smokers, which is oxidant primed, making it more sensitive to bacterial or PAMP stimulation when compared with blood of nonsmokers. Thus an oxidant stimulation, possibly via an action on TLR2 or associated transduction pathways, provides a signal that initiates inflammatory responses and sensitizes cells to pathogenic insults.


Subject(s)
Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/physiology , Interleukin-8/metabolism , Monocytes/drug effects , Oxidants/pharmacology , Cell Line , Cell Respiration/drug effects , Diglycerides/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Immunity, Innate , Interleukin-8/genetics , Lipopeptides , Lipopolysaccharides/pharmacology , Models, Biological , Monocytes/metabolism , Monocytes/microbiology , Nod1 Signaling Adaptor Protein/agonists , Oligopeptides/pharmacology , Peptides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Smoking/blood , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 6/antagonists & inhibitors
20.
J Endocrinol ; 193(3): 323-30, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17535871

ABSTRACT

Pathogens are sensed by pattern recognition receptors (PRRs), which are germ line-encoded receptors, including transmembrane Toll-like receptors (TLRs) and cytosolic nucleotide oligomerisation domain (NOD) proteins, containing leucine-rich repeats (NLRs). Activation of PRRs by specific pathogen-associated molecular patterns (PAMPs) results in genomic responses in host cells involving activation transcription factors and the induction of genes. There are now at least 10 TLRs in humans and 13 in mice, and 2 NLRs (NOD1 and NOD2). TLR signalling is via interactions with adaptor proteins including MyD88 and toll-receptor associated activator of interferon (TRIF). NOD signalling is via the inflammasome and involves activation of Rip-like interactive clarp kinase (RICK). Bacterial lipopolysaccharide (LPS) from Gram-negative bacteria is the best-studied PAMP and is activated by or 'sensed' by TLR4. Lipoteichoic acid (LTA) from Gram-positive bacteria is sensed by TLR2. TLR4 and TLR2 have different signalling cascades, although activation of either results in symptoms of sepsis and shock. This review describes the rapidly expanding field of pathogen-sensing receptors and uses LPS and LTA as examples of how these pathways parallel and diverge from each other. The role of pathogen-sensing pathways in disease is also discussed.


Subject(s)
Bacterial Infections/immunology , Nod Signaling Adaptor Proteins/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Bacterial Infections/metabolism , Humans , Lipopolysaccharides/metabolism , Mice , Viral Proteins/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...