Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
FASEB J ; 31(6): 2439-2445, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28246166

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Interleucina-1beta/metabolismo , Monocitos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular Tumoral , Conexinas/genética , Diglicéridos/farmacología , Regulación de la Expresión Génica/fisiología , Humanos , Interleucina-1beta/genética , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Proteínas del Tejido Nervioso/genética , Oligopéptidos/farmacología , Receptores Purinérgicos P2X7/genética , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-23742950

RESUMEN

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.


Asunto(s)
Ciclooxigenasa 1/genética , Ciclooxigenasa 2/genética , Citocinas/metabolismo , Fibroblastos/enzimología , Proteínas de la Membrana/genética , Receptores Toll-Like/agonistas , Animales , Proliferación Celular , Células Cultivadas , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Diclofenaco/farmacología , Dinoprostona/metabolismo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Interacciones Huésped-Patógeno , Humanos , Fibrosis Pulmonar Idiopática/enzimología , Inmunidad Innata , Lipopolisacáridos/farmacología , Pulmón/inmunología , Pulmón/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/farmacología , Receptores Toll-Like/metabolismo
3.
Biochem Biophys Res Commun ; 426(4): 486-91, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22960172

RESUMEN

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.


Asunto(s)
Endotelina-1/biosíntesis , Hipertensión Pulmonar/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor Toll-Like 3/metabolismo , Células Cultivadas , Quimiocinas/genética , Citocinas/genética , Hipertensión Pulmonar Primaria Familiar , Expresión Génica , Humanos , Hipertensión Pulmonar/virología , Interleucina-8/genética , Músculo Liso Vascular/virología , Miocitos del Músculo Liso/virología , Poli I-C/farmacología , Receptores de Citocinas/genética , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/genética , Factor de Necrosis Tumoral alfa/farmacología
4.
Am J Respir Crit Care Med ; 184(11): 1261-9, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21868501

RESUMEN

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.


Asunto(s)
Endotoxemia/microbiología , Cardiopatías Congénitas/cirugía , Enfermedades Intestinales/microbiología , Mucosa Intestinal/lesiones , Mucosa Intestinal/microbiología , Regulación hacia Abajo/inmunología , Endotoxemia/sangre , Endotoxemia/inmunología , Ensayo de Inmunoadsorción Enzimática , Proteínas de Unión a Ácidos Grasos/sangre , Proteínas de Unión a Ácidos Grasos/inmunología , Femenino , Humanos , Lactante , Inflamación/sangre , Inflamación/inmunología , Inflamación/microbiología , Enfermedades Intestinales/sangre , Enfermedades Intestinales/inmunología , Mucosa Intestinal/inmunología , Tiempo de Internación/estadística & datos numéricos , Masculino , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/microbiología , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/microbiología , Índice de Severidad de la Enfermedad
5.
Front Pharmacol ; 13: 887608, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35814225

RESUMEN

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.

6.
J Exp Med ; 200(1): 69-78, 2004 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15238606

RESUMEN

The established model for the mechanism of action of aspirin is the inhibition of prostaglandin synthesis. However, this has never fully explained aspirin's repertoire of antiinflammatory properties. We found in acute pleuritis that aspirin, but not salicylate, indomethacin, or piroxicam, increased plasma nitric oxide (NO), which correlated with a reduction in inflammation. Inhibiting aspirin-elicited NO pharmacologically in this model nullified the antiinflammatory effects of aspirin. Moreover, aspirin was not antiinflammatory in either constitutive (eNOS) or inducible NO synthase (iNOS) knockout mice with IL-1beta-induced peritonitis. It transpires that aspirin generates NO through its unique ability to trigger the synthesis of 15-epi-lipoxin A(4). Aspirin and 15-epi-lipoxin A(4) were shown to inhibit leukocyte trafficking in an NO-dependent manner using intravital microscopy on IL-1beta-stimulated mouse mesentery. Not only did aspirin inhibit leukocyte-endothelial interaction in a manner similar to NO in wild-type mice but both aspirin and 15-epi-lipoxin A(4) had markedly reduced effects on leukocyte-endothelial cell adherence in eNOS- and iNOS-deficient mice compared with wild type. Collectively, these data suggest that aspirin triggers the synthesis of 15-epi-lipoxin A(4), which increases NO synthesis through eNOS and iNOS. This aspirin-elicited NO exerts antiinflammatory effects in the microcirculation by inhibiting leukocyte-endothelium interactions.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Aspirina/metabolismo , Inflamación/metabolismo , Lipoxinas/metabolismo , Óxido Nítrico/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Aspirina/uso terapéutico , Relación Dosis-Respuesta a Droga , Endotelio/metabolismo , Inflamación/tratamiento farmacológico , Interleucina-1/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Lipoxinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Nitritos/sangre , Peritonitis/metabolismo , Ratas , Ratas Wistar
7.
Arterioscler Thromb Vasc Biol ; 29(5): 706-11, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19150877

RESUMEN

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.


Asunto(s)
Plaquetas/efectos de los fármacos , Ácido Clofíbrico/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipolipemiantes/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Animales , Plaquetas/fisiología , Humanos , Ratones , Ratones Noqueados , PPAR alfa/efectos de los fármacos , PPAR gamma/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos
8.
Am J Respir Crit Care Med ; 179(4): 299-306, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19011150

RESUMEN

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.


Asunto(s)
Bronquitis/metabolismo , Oxidantes/metabolismo , Peritonitis/metabolismo , Receptor Toll-Like 2/inmunología , Animales , Western Blotting/métodos , Bronquitis/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Ratones , Ratones Endogámicos C57BL , Oxidantes/inmunología , Estrés Oxidativo , Peritonitis/inmunología , Fumar/inmunología , Fumar/metabolismo
10.
Sci Rep ; 9(1): 9218, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31239465

RESUMEN

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.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Fibras Musculares de Contracción Rápida/efectos de los fármacos , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Fibras Musculares de Contracción Lenta/metabolismo , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Anciano , Animales , Estudios de Casos y Controles , Factor de Crecimiento Epidérmico/genética , Femenino , Humanos , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Persona de Mediana Edad , Fibras Musculares de Contracción Rápida/fisiología , Fibras Musculares de Contracción Lenta/fisiología , Oxidación-Reducción/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , ARN Mensajero/genética , Pez Cebra
11.
Sci Rep ; 8(1): 13987, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30228360

RESUMEN

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.


Asunto(s)
Aminas/sangre , Aminoácidos/sangre , Arginina/análogos & derivados , Biomarcadores/sangre , Cromatografía Líquida de Alta Presión/métodos , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Espectrometría de Masas en Tándem/métodos , Anciano , Arginina/sangre , Femenino , Humanos , Masculino , Pronóstico , Síndrome de Respuesta Inflamatoria Sistémica/cirugía
12.
J Endocrinol ; 193(3): 323-30, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17535871

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Proteínas Adaptadoras de Señalización NOD/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Animales , Infecciones Bacterianas/metabolismo , Humanos , Lipopolisacáridos/metabolismo , Ratones , Proteínas Virales/metabolismo , Virosis/inmunología , Virosis/metabolismo
13.
Shock ; 27(1): 40-7, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17172979

RESUMEN

Pathogens contain specific pathogen-associated molecular patterns, which activate pattern recognition receptors of the innate immune system such as Toll-like receptors (TLRs). Although there is a clear evidence of how macrophages sense pathogens, we know less about such processes in vessels. This is critical to understand because activation of vascular cells and the subsequent induction of inflammatory genes by bacteria are crucial events in the development of septic shock. In the current study we have used genetically modified mice to investigate the role of TLRs, adapter proteins, tumor necrosis factor alpha (TNFalpha), and nitric oxide synthase II (NOSII) in vascular dysfunction induced by Gram-positive (Staphylococcus aureus) or Gram-negative (Escherichia coli) bacteria. Our data show that Gram-positive S. aureus or Gram-negative E. coli causes vascular dysfunction via the induction of NOSII. For S. aureus, this process requires TLR2, TLR6, myeloid differentiation factor 88 (MyD88) adapter-like, MyD88, and TNF, but not TLR4 or TLR1. Vascular dysfunction induced by E. coli requires TLR4 but has no requirement for TLR2, TLR1, TLR6, or TNF, and a partial but incomplete requirement of MyD88 and TIR domain-containing adapter inducing interferon-beta. Staphylococcus aureus induced NOSII protein expression in vascular smooth muscle cells but not in macrophages, whereas E. coli induced NOSII in both cell types. Our data are the first to establish the definitive roles of specific TLRs in the sensing of Gram-positive and Gram-negative bacteria by vessels and demonstrate that macrophages and blood vessels may differ in their response to pathogens.


Asunto(s)
Escherichia coli/fisiología , Transducción de Señal/fisiología , Staphylococcus aureus/fisiología , Receptores Toll-Like/fisiología , Enfermedades Vasculares/patología , Animales , Células Cultivadas , Macrófagos/enzimología , Ratones , Ratones Noqueados , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Ratas , Enfermedades Vasculares/microbiología
14.
FASEB J ; 20(2): 326-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16368717

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that heterodimerize with the retinoid X receptor and then modulate at the transcriptional level the function of many target genes. Three PPARs are known: alpha, beta (sometimes called delta), and gamma. The better studied are PPARalpha and PPARgamma, which are activated by fibrates and thiazolidinediones/glitazones, respectively. It is now believed that activation of the PPARs could be associated with the prevention of heart attack and stroke in humans. Here we report, for the first time, that human platelets contain PPARbeta and that its selective activation inhibits platelet aggregation. PPARbeta is a putative receptor for prostacyclin. Prostacyclin is an important antithrombotic hormone that synergizes with nitric oxide to inhibit platelet aggregation. In the current study, we show that PPARbeta ligands similarly synergize with nitric oxide to inhibit platelet aggregation. These observations challenge our view of a nuclear receptor because PPARbeta is present and active in nonnucleated platelets. Furthermore, these data suggest that some of the antithrombotic actions of prostacyclin may be mediated via activation of PPARs. Thus, our results identify PPARbeta as a novel antiplatelet target that may mediate some of the effects of prostacyclin in blood.


Asunto(s)
Plaquetas/metabolismo , PPAR-beta/metabolismo , Transducción de Señal , Calcio/metabolismo , Línea Celular , Humanos , Megacariocitos/metabolismo , Óxido Nítrico/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología
15.
PLoS One ; 12(8): e0183025, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28813479

RESUMEN

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.


Asunto(s)
Aminas/metabolismo , Metabolómica , Sepsis/metabolismo , Adulto , Anciano , Animales , Arginina/metabolismo , Línea Celular , Cromatografía Liquida , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Espectrometría de Masas , Ratones , Persona de Mediana Edad , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Sepsis/fisiopatología , omega-N-Metilarginina/metabolismo
16.
Br J Pharmacol ; 148(8): 1067-75, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16783405

RESUMEN

1. Gram-negative and Gram-positive bacteria are sensed by Toll-like receptor (TLR)4 and TLR2, respectively. TLR4 recruits MyD88 and TRIF, whereas TLR2 recruits MyD88 without TRIF. NOSII and TNFalpha are central genes in innate immunity and are thought to be differentially regulated by the MyD88 versus TRIF signalling pathways. Here, we have used Gram-positive Staphylococcus aureus, Gram-negative Escherichia coli and highly selective TLR ligands to establish the precise relationship between TLR2, TLR1, TLR6 and TLR4 for NOSII versus TNFalpha induction. 2. In murine macrophages at 24 h, E. coli or LPS (TLR4) induced NO and TNFalpha release. In contrast, S. aureus (TLR2/TLR1/TLR6) or Pam(3)CSK4 (TLR2/TLR1), or FSL-1 and LTA (TLR2/TLR6) induced TNFalpha without an effect on NO. 3. At later time points (48-72 h), S. aureus induced NO release. The ability of S. aureus, but not E. coli or LPS, to induce NO release was inhibited by anti-TNFalpha-binding antibodies. 4. At 24 h, LPS synergised with TLR2 ligands to induce NO release and NOSII protein expression. LPS also induced the expression of TLR2 gene expression without affecting levels of TLR4. 5. Using cells from TLR2(-/-) or TLR4(-/-) mice, the ability of LPS to synergise with S. aureus or Pam(3)CSK4 was found to be dependent on both TLR2 and TLR4. 6. These observations are the first to clearly delineate the role of separately activating TLR2 and TLR4 in the induction of NOSII and TNFalpha genes compared with their coinduction when both receptor pathways are activated.


Asunto(s)
Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/fisiología , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , Activación de Macrófagos , Macrófagos/enzimología , Macrófagos/microbiología , Ratones , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
17.
PLoS One ; 11(2): e0147289, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26910063

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Carcinogénesis/efectos de los fármacos , Sulfuro de Hidrógeno/química , Neoplasias Intestinales/patología , Neoplasias Intestinales/prevención & control , Naproxeno/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/uso terapéutico , Quimioprevención , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Pólipos Intestinales/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Naproxeno/química , Naproxeno/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/metabolismo
18.
FASEB J ; 17(2): 253-5, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12475898

RESUMEN

The 37-kDa protein annexin 1 (Anx-1; lipocortin 1) has been implicated in the regulation of phagocytosis, cell signaling, and proliferation and is postulated to be a mediator of glucocorticoid action in inflammation and in the control of anterior pituitary hormone release. Here, we report that mice lacking the Anx-1 gene exhibit a complex phenotype that includes an altered expression of other annexins as well as of COX-2 and cPLA2. In carrageenin- or zymosan-induced inflammation, Anx-1-/- mice exhibit an exaggerated response to the stimuli characterized by an increase in leukocyte emigration and IL-1beta generation and a partial or complete resistance to the antiinflammatory effects of glucocorticoids. Anx-1-/- polymorphonuclear leucocytes exhibited increased spontaneous migratory behavior in vivo whereas in vitro, leukocytes from Anx-1-/- mice had reduced cell surface CD 11b (MAC-1) but enhanced CD62L (L-selectin) expression and Anx-1-/- macrophages exhibited anomalies in phagocytosis. There are also gender differences in activated leukocyte behavior in the Anx-1-/- mice that are not seen in the wild-type animals, suggesting an interaction between sex hormones and inflammation in Anx-1-/- animals.


Asunto(s)
Anexina A1/genética , Dexametasona/farmacología , Glucocorticoides/farmacología , Inflamación/tratamiento farmacológico , Animales , Anexina A1/metabolismo , Carragenina , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Ciclooxigenasa 2 , Resistencia a Medicamentos , Edema/inducido químicamente , Edema/tratamiento farmacológico , Edema/genética , Femenino , Genotipo , Inflamación/inducido químicamente , Inflamación/genética , Interleucina-1/metabolismo , Isoenzimas/efectos de los fármacos , Isoenzimas/metabolismo , Leucocitos/citología , Masculino , Ratones , Ratones Noqueados , Fosfolipasas A/efectos de los fármacos , Fosfolipasas A/metabolismo , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/metabolismo , Factores Sexuales
19.
Biochem J ; 376(Pt 1): 285-90, 2003 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12948397

RESUMEN

Glucocorticoids inhibit the release of eicosanoid pro-inflammatory mediators. The immunosuppressant FK506 is known to enhance many aspects of glucocorticoid action. In the present study we show that FK506 (1 microM or 10 microM) inhibits the release of arachidonic acid and prostaglandin E2 from A549 cells and also inhibits their proliferation. Simultaneous treatment of FK506 together with the glucocorticoids dexamethasone, methyl-prednisolone, fluticasone or mometasone (10 nM) enhances the growth inhibitory effect of these steroids. Furthermore, the simultaneous use of FK506 and these glucocorticoids similarly results in enhanced inhibition of arachidonic acid release. When pretreated for 2 h, FK506 enhances glucocorticoid inhibition of COX2 (cyclo-oxygenase 2) expression. However, when administered simultaneously, FK506 blocks glucocorticoid inhibition of COX2 expression. Nuclear uptake of glucocorticoid receptors mediated by glucocorticoids is also blocked by the simultaneous administration of FK506. These results suggest that the effect of simultaneous treatment of FK506 with glucocorticoids differs significantly from that where pre-treatment of the immunosuppressant is used. Recently, immunophilin interchange has been identified as a first step in glucocorticoid receptor activation following ligand activation. We show here that the FKB51 (FK506-binding protein 51)-FKB52 switch is differentially regulated by glucocorticoid and FK506 treatment strategy.


Asunto(s)
Glucocorticoides/farmacología , Inmunosupresores/farmacología , Tacrolimus/farmacología , Ácido Araquidónico/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclooxigenasa 2 , Dinoprostona/biosíntesis , Sinergismo Farmacológico , Glucocorticoides/antagonistas & inhibidores , Humanos , Isoenzimas/metabolismo , Proteínas de la Membrana , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
20.
BMJ Open ; 5(1): e005750, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25631307

RESUMEN

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.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Proyectos de Investigación , Fumar/inmunología , Fumar/metabolismo , Administración por Inhalación , Humanos , Modelos Biológicos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA