Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 52
Filtrer
Plus de filtres











Gamme d'année
2.
Front Pharmacol ; 13: 887608, 2022.
Article de Anglais | MEDLINE | ID: mdl-35814225

RÉSUMÉ

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.
Preprint de Anglais | bioRxiv | ID: ppbiorxiv-484817

RÉSUMÉ

The Omicron variant (B.1.1.529) of SARS-CoV-2 rapidly becomes dominant globally. Its extensive mutations confer severe efficacy reduction to most of existing antibodies or vaccines. Here, we developed RAMIHM, a highly efficient strategy to generate fully human monoclonal antibodies (mAbs), directly applied it with Omicron-mRNA immunization, and isolated three potent and specific clones against Omicron. Rapid mRNA immunization elicited strong anti-Omicron antibody response in humanized mice, along with broader anti-coronavirus activity. Customized single cell BCR sequencing mapped the clonal repertoires. Top-ranked clones collectively from peripheral blood, plasma B and memory B cell populations showed high rate of Omicron-specificity (93.3%) from RAMIHM-scBCRseq. Clone-screening identified three highly potent neutralizing antibodies that have low nanomolar affinity for Omicron RBD, and low ng/mL level IC50 in neutralization, more potent than majority of currently approved or authorized clinical RBD-targeting mAbs. These lead mAbs are fully human and ready for downstream IND-enabling and/or translational studies.

4.
Preprint de Anglais | bioRxiv | ID: ppbiorxiv-480449

RÉSUMÉ

The Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has high transmissibility and recently swept the globe. Due to the extensive number of mutations, this variant has high level of immune evasion, which drastically reduced the efficacy of existing antibodies and vaccines. Thus, it is important to test an Omicron-specific vaccine, evaluate its immune response against Omicron and other variants, and compare its immunogenicity as boosters with existing vaccine designed against the reference wildtype virus (WT). Here, we generated an Omicron-specific lipid nanoparticle (LNP) mRNA vaccine candidate, and tested its activity in animals, both alone and as a heterologous booster to existing WT mRNA vaccine. Our Omicron-specific LNP-mRNA vaccine elicited strong and specific antibody response in vaccination-naive mice. Mice that received two-dose WT LNP-mRNA, the one mimicking the commonly used Pfizer/Moderna mRNA vaccine, showed a >40-fold reduction in neutralization potency against Omicron variant than that against WT two weeks post second dose, which further reduced to background level >3 months post second dose. As a booster shot for two-dose WT mRNA vaccinated mice, a single dose of either a homologous booster with WT LNP-mRNA or a heterologous booster with Omicron LNP-mRNA restored the waning antibody response against Omicron, with over 40-fold increase at two weeks post injection as compared to right before booster. Interestingly, the heterologous Omicron LNP-mRNA booster elicited neutralizing titers 10-20 fold higher than the homologous WT booster against the Omicron variant, with comparable titers against the Delta variant. All three types of vaccination, including Omicron mRNA alone, WT mRNA homologous booster, and Omicron heterologous booster, elicited broad binding antibody responses against SARS-CoV-2 WA-1, Beta, and Delta variants, as well as other Betacoronavirus species such as SARS-CoV, but not Middle East respiratory syndrome coronavirus (MERS-CoV). These data provided direct proof-of-concept assessments of an Omicron-specific mRNA vaccination in vivo, both alone and as a heterologous booster to the existing widely-used WT mRNA vaccine form.

5.
Preprint de Anglais | bioRxiv | ID: ppbiorxiv-473733

RÉSUMÉ

COVID-19 pathogen SARS-CoV-2 has infected hundreds of millions and caused over 5 million deaths to date. Although multiple vaccines are available, breakthrough infections occur especially by emerging variants. Effective therapeutic options such as monoclonal antibodies (mAbs) are still critical. Here, we report the development, cryo-EM structures, and functional analyses of mAbs that potently neutralize SARS-CoV-2 variants of concern. By high-throughput single cell sequencing of B cells from spike receptor binding domain (RBD) immunized animals, we identified two highly potent SARS-CoV-2 neutralizing mAb clones that have single-digit nanomolar affinity and low-picomolar avidity, and generated a bispecific antibody. Lead antibodies showed strong inhibitory activity against historical SARS-CoV-2 and several emerging variants of concern. We solved several cryo-EM structures at [~]3 [A] resolution of these neutralizing antibodies in complex with prefusion spike trimer ectodomain, and revealed distinct epitopes, binding patterns, and conformations. The lead clones also showed potent efficacy in vivo against authentic SARS-CoV-2 in both prophylactic and therapeutic settings. We also generated and characterized a humanized antibody to facilitate translation and drug development. The humanized clone also has strong potency against both the original virus and the B.1.617.2 Delta variant. These mAbs expand the repertoire of therapeutics against SARS-CoV-2 and emerging variants.

6.
Sci Rep ; 9(1): 9218, 2019 06 25.
Article de Anglais | MEDLINE | ID: mdl-31239465

RÉSUMÉ

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.


Sujet(s)
Récepteurs ErbB/antagonistes et inhibiteurs , Fibres musculaires à contraction rapide/effets des médicaments et des substances chimiques , Fibres musculaires à contraction rapide/métabolisme , Fibres musculaires à contraction lente/effets des médicaments et des substances chimiques , Fibres musculaires à contraction lente/métabolisme , Phénotype , Inhibiteurs de protéines kinases/pharmacologie , Sujet âgé , Animaux , Études cas-témoins , Facteur de croissance épidermique/génétique , Femelle , Humains , Locomotion/effets des médicaments et des substances chimiques , Locomotion/physiologie , Mâle , Souris , Adulte d'âge moyen , Fibres musculaires à contraction rapide/physiologie , Fibres musculaires à contraction lente/physiologie , Oxydoréduction/effets des médicaments et des substances chimiques , Broncho-pneumopathie chronique obstructive/génétique , Broncho-pneumopathie chronique obstructive/physiopathologie , ARN messager/génétique , Danio zébré
7.
Sci Rep ; 8(1): 13987, 2018 09 18.
Article de Anglais | MEDLINE | ID: mdl-30228360

RÉSUMÉ

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.


Sujet(s)
Amines/sang , Acides aminés/sang , Arginine/analogues et dérivés , Marqueurs biologiques/sang , Chromatographie en phase liquide à haute performance/méthodes , Syndrome de réponse inflammatoire généralisée/sang , Spectrométrie de masse en tandem/méthodes , Sujet âgé , Arginine/sang , Femelle , Humains , Mâle , Pronostic , Syndrome de réponse inflammatoire généralisée/chirurgie
8.
PLoS One ; 12(8): e0183025, 2017.
Article de Anglais | MEDLINE | ID: mdl-28813479

RÉSUMÉ

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.


Sujet(s)
Amines/métabolisme , Métabolomique , Sepsie/métabolisme , Adulte , Sujet âgé , Animaux , Arginine/métabolisme , Lignée cellulaire , Chromatographie en phase liquide , Test ELISA , Femelle , Humains , Mâle , Spectrométrie de masse , Souris , Adulte d'âge moyen , Nitric oxide synthase/métabolisme , Nitric oxide synthase type II/métabolisme , Sepsie/physiopathologie , oméga-N-Méthylarginine/métabolisme
9.
FASEB J ; 31(6): 2439-2445, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28246166

RÉSUMÉ

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.


Sujet(s)
Adénosine triphosphate/métabolisme , Connexines/métabolisme , Interleukine-1 bêta/métabolisme , Monocytes/métabolisme , Protéines de tissu nerveux/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Adénosine triphosphate/génétique , Caspase-1/génétique , Caspase-1/métabolisme , Lignée cellulaire tumorale , Connexines/génétique , Diglycéride/pharmacologie , Régulation de l'expression des gènes/physiologie , Humains , Interleukine-1 bêta/génétique , Lipopeptides/pharmacologie , Lipopolysaccharides/pharmacologie , Protéines de tissu nerveux/génétique , Oligopeptides/pharmacologie , Récepteurs purinergiques P2X7/génétique , Récepteur de type Toll-2/agonistes , Récepteur de type Toll-2/génétique , Récepteur de type Toll-2/métabolisme , Récepteur de type Toll-4/agonistes , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme
10.
PLoS One ; 11(2): e0147289, 2016.
Article de Anglais | MEDLINE | ID: mdl-26910063

RÉSUMÉ

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.


Sujet(s)
Protéine de la polypose adénomateuse colique/génétique , Carcinogenèse/effets des médicaments et des substances chimiques , Sulfure d'hydrogène/composition chimique , Tumeurs de l'intestin/anatomopathologie , Tumeurs de l'intestin/prévention et contrôle , Naproxène/analogues et dérivés , Animaux , Anti-inflammatoires non stéroïdiens/composition chimique , Anti-inflammatoires non stéroïdiens/usage thérapeutique , Chimioprévention , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Côlon/anatomopathologie , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Polypes intestinaux/prévention et contrôle , Mâle , Souris , Souris de lignée C57BL , Naproxène/composition chimique , Naproxène/usage thérapeutique , Protéines proto-oncogènes c-myc/métabolisme , bêta-Caténine/métabolisme
11.
BMJ Open ; 5(1): e005750, 2015 Jan 28.
Article de Anglais | MEDLINE | ID: mdl-25631307

RÉSUMÉ

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.


Sujet(s)
Broncho-pneumopathie chronique obstructive/immunologie , Broncho-pneumopathie chronique obstructive/métabolisme , Plan de recherche , Fumer/immunologie , Fumer/métabolisme , Administration par inhalation , Humains , Modèles biologiques , Muqueuse nasale/effets des médicaments et des substances chimiques , Muqueuse nasale/immunologie , Muqueuse nasale/métabolisme , Emphysème pulmonaire/immunologie , Emphysème pulmonaire/métabolisme
12.
PLoS One ; 9(6): e98165, 2014.
Article de Anglais | MEDLINE | ID: mdl-24887395

RÉSUMÉ

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.


Sujet(s)
Athérosclérose/enzymologie , Vaisseaux sanguins/métabolisme , Cyclooxygenase 2/métabolisme , Prostacycline/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Agents protecteurs/métabolisme , Lymphocytes T/métabolisme , Animaux , Apolipoprotéines E/déficit , Apolipoprotéines E/métabolisme , Athérosclérose/anatomopathologie , Vaisseaux sanguins/anatomopathologie , Cyclooxygenase 2/déficit , Cyclooxygenase 2/génétique , Femelle , Délétion de gène , Régulation de l'expression des gènes codant pour des enzymes , Mâle , Souris de lignée C57BL , Transduction du signal , Transcriptome/génétique , Régulation positive/génétique
13.
Article de Anglais | MEDLINE | ID: mdl-23742950

RÉSUMÉ

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.


Sujet(s)
Cyclooxygenase 1/génétique , Cyclooxygenase 2/génétique , Cytokines/métabolisme , Fibroblastes/enzymologie , Protéines membranaires/génétique , Récepteurs de type Toll/agonistes , Animaux , Prolifération cellulaire , Cellules cultivées , Cyclooxygenase 1/métabolisme , Cyclooxygenase 2/métabolisme , Inhibiteurs des cyclooxygénases/pharmacologie , Diclofenac/pharmacologie , Dinoprostone/métabolisme , Fibroblastes/immunologie , Fibroblastes/métabolisme , Interactions hôte-pathogène , Humains , Fibrose pulmonaire idiopathique/enzymologie , Immunité innée , Lipopolysaccharides/pharmacologie , Poumon/immunologie , Poumon/anatomopathologie , Protéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Poly I-C/pharmacologie , Récepteurs de type Toll/métabolisme
14.
Pulm Circ ; 3(4): 926-35, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-25006409

RÉSUMÉ

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.

15.
Biochem Biophys Res Commun ; 426(4): 486-91, 2012 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-22960172

RÉSUMÉ

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.


Sujet(s)
Endothéline-1/biosynthèse , Hypertension pulmonaire/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Récepteur de type Toll-3/métabolisme , Cellules cultivées , Chimiokines/génétique , Cytokines/génétique , Hypertension artérielle pulmonaire primitive familiale , Expression des gènes , Humains , Hypertension pulmonaire/virologie , Interleukine-8/génétique , Muscles lisses vasculaires/virologie , Myocytes du muscle lisse/virologie , Poly I-C/pharmacologie , Récepteurs aux cytokines/génétique , Récepteur de type Toll-3/agonistes , Récepteur de type Toll-3/génétique , Facteur de nécrose tumorale alpha/pharmacologie
17.
Pharmacol Ther ; 135(2): 200-15, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22627269

RÉSUMÉ

Pharmacologists have used pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS) for decades as a stimulus for studying mediators involved in inflammation and for the screening of anti-inflammatory compounds. However, in the view of immunologists, LPS was too non-specific for studying the mechanisms of immune signalling in infection and inflammation, as no receptors had been identified. This changed in the late 1990s with the discovery of the Toll-like receptors. These 'pattern recognition receptors' (PRRs) were able to recognise highly conserved sequences, the so called pathogen associated molecular patterns (PAMPs) present in or on pathogens. This specificity of particular PAMPs and their newly defined receptors provided a common ground between pharmacologists and immunologists for the study of inflammation. PRRs also recognise endogenous agonists, the so called danger-associated molecular patterns (DAMPs), which can result in sterile inflammation. The signalling pathways and ligands of many PRRs have now been characterised and there is no doubt that this rich vein of research will aid the discovery of new therapeutics for infectious conditions and chronic inflammatory disease.


Sujet(s)
Inflammation/immunologie , Récepteurs de reconnaissance de motifs moléculaires/immunologie , Animaux , Humains , Récepteurs de type Toll/immunologie
18.
PLoS One ; 7(2): e30120, 2012.
Article de Anglais | MEDLINE | ID: mdl-22363418

RÉSUMÉ

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.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Inflammation/génétique , Monocytes/métabolisme , Monocytes/anatomopathologie , Fumer/génétique , Lignée cellulaire , Bases de données comme sujet , Régulation de l'expression des gènes , Réseaux de régulation génique/génétique , Heme oxygenase-1/génétique , Heme oxygenase-1/métabolisme , Humains , Interleukine-8/génétique , Interleukine-8/métabolisme , Analyse en composantes principales , Transduction du signal/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme
19.
Am J Respir Crit Care Med ; 184(11): 1261-9, 2011 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-21868501

RÉSUMÉ

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.


Sujet(s)
Endotoxémie/microbiologie , Cardiopathies congénitales/chirurgie , Maladies intestinales/microbiologie , Muqueuse intestinale/traumatismes , Muqueuse intestinale/microbiologie , Régulation négative/immunologie , Endotoxémie/sang , Endotoxémie/immunologie , Test ELISA , Protéines de liaison aux acides gras/sang , Protéines de liaison aux acides gras/immunologie , Femelle , Humains , Nourrisson , Inflammation/sang , Inflammation/immunologie , Inflammation/microbiologie , Maladies intestinales/sang , Maladies intestinales/immunologie , Muqueuse intestinale/immunologie , Durée du séjour/statistiques et données numériques , Mâle , Défaillance multiviscérale/sang , Défaillance multiviscérale/immunologie , Défaillance multiviscérale/microbiologie , Complications postopératoires/sang , Complications postopératoires/immunologie , Complications postopératoires/microbiologie , Indice de gravité de la maladie
20.
Br J Pharmacol ; 160(8): 1997-2007, 2010 Aug.
Article de Anglais | MEDLINE | ID: mdl-20649597

RÉSUMÉ

BACKGROUND AND PURPOSE: Gram-negative bacteria contain ligands for Toll-like receptor (TLR) 4 and nucleotide oligomerization domain (NOD) 1 receptors. Lipopolysaccharide (LPS) activates TLR4, while peptidoglycan products activate NOD1. Activation of NOD1 by the specific agonist FK565 results in a profound vascular dysfunction and experimental shock in vivo. EXPERIMENTAL APPROACH: Here, we have analysed a number of pharmacological inhibitors to characterize the role of key signalling pathways in the induction of NOS2 following TLR4 or NOD1 activation. KEY RESULTS: Vascular smooth muscle (VSM) cells expressed NOD1 mRNA and protein, and, after challenge with Escherichia coli or FK565, NOS2 protein and activity were induced. Macrophages had negligible levels of NOD1 and were unaffected by FK565, but responded to E. coli and LPS by releasing increased NO and expression of NOS2 protein. Classic pharmacological inhibitors for NF-kappaB (SC-514) and mitogen-activated protein kinase (SB203580, PD98059) signalling pathways inhibited responses in both cell types regardless of agonist. While TLR4-mediated responses in macrophages were specifically inhibited by the pan-caspase inhibitor z-VAD-fmk and the PKC inhibitor Gö6976, NOD1-mediated responses in VSM cells were inhibited by the Rip2 inhibitor PP2. CONCLUSIONS AND IMPLICATIONS: Our findings suggest a selective role for NOD1 in VSM cells, and highlight NOD1 as a potential novel therapeutic target for the treatment of vascular inflammation.


Sujet(s)
Macrophages péritonéaux/enzymologie , Muscles lisses vasculaires/enzymologie , Myocytes du muscle lisse/enzymologie , Nitric oxide synthase type II/biosynthèse , Protéine adaptatrice de signalisation NOD1/métabolisme , Récepteur de type Toll-4/métabolisme , Animaux , Lignée cellulaire , Relation dose-effet des médicaments , Induction enzymatique , Médiateurs de l'inflammation/métabolisme , Lipopolysaccharides/pharmacologie , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/immunologie , Souris , Souris de lignée C57BL , Mitogen-Activated Protein Kinases/métabolisme , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Monoxyde d'azote/métabolisme , Protéine adaptatrice de signalisation NOD1/agonistes , Protéine adaptatrice de signalisation NOD1/génétique , Oligopeptides/pharmacologie , Protéine kinase C/métabolisme , ARN messager/métabolisme , Rats , Rats de lignée WKY , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/agonistes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE