Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1085-L1092, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33822656

RESUMEN

Resolution of the acute respiratory distress syndrome (ARDS) from pneumonia requires repair of the injured lung endothelium and alveolar epithelium, removal of neutrophils from the distal airspaces of the lung, and clearance of the pathogen. Previous studies have demonstrated the importance of specialized proresolving mediators (SPMs) in the regulation of host responses during inflammation. Although ARDS is commonly caused by Streptococcus pneumoniae, the role of lipoxin A4 (LXA4) and resolvin D1 (RvD1) in pneumococcal pneumonia is not well understood. In the present experimental study, we tested the hypothesis that endogenous SPMs play a role in the resolution of lung injury in a clinically relevant model of bacterial pneumonia. Blockade of formyl peptide receptor 2 (ALX/FPR2), the receptor for LXA4 and RvD1, with the peptide WRW4 resulted in more pulmonary edema, greater protein accumulation in the air spaces, and increased bacteria accumulation in the air spaces and the blood. Inhibition of this receptor was also associated with decreased levels of proinflammatory cytokines. Even in the presence of antibiotic treatment, WRW4 inhibited the resolution of lung injury. In summary, these experiments demonstrated two novel findings: LXA4 and RvD1 contribute to the resolution of lung injury due to pneumococcal pneumonia, and the mechanism of their benefit likely includes augmenting bacterial clearance and reducing pulmonary edema via the restoration of lung alveolar-capillary barrier permeability.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Ácidos Docosahexaenoicos/antagonistas & inhibidores , Lipoxinas/antagonistas & inhibidores , Neumonía Neumocócica/tratamiento farmacológico , Receptores de Lipoxina/efectos de los fármacos , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/inmunología , Animales , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Permeabilidad/efectos de los fármacos , Neumonía Neumocócica/complicaciones , Neumonía Neumocócica/inmunología , Receptores de Lipoxina/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/inmunología
2.
Arthritis Rheumatol ; 69(5): 996-1008, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27860453

RESUMEN

OBJECTIVE: Pain is a major symptom of osteoarthritis (OA); currently available analgesics either do not provide adequate pain relief or are associated with serious side effects. The aim of this study was to investigate the therapeutic potential of targeting the resolvin receptor system to modify OA pain and pathology. METHODS: Gene expression of 2 resolvin receptors (ALX and ChemR23) was quantified in synovium and medial tibial plateau specimens obtained from patients with OA at the time of joint replacement surgery. Two models of OA joint pain were used for the mechanistic studies. Gene expression in the joint and central nervous system was quantified. The effects of exogenous administration of the D series resolvin precursor 17(R)-hydroxy-docosahexaenoic acid (17[R]-HDoHE) on pain behavior, joint pathology, spinal microglia, and astroglyosis were quantified. Plasma levels of relevant lipids, resolvin D2, 17(R)-HDoHE, and arachidonic acid, were determined in rats, using liquid chromatography tandem mass spectrometry. RESULTS: There was a positive correlation between resolvin receptor and interleukin-6 (IL-6) expression in human OA synovial and medial tibial plateau tissue. In rats, synovial expression of ALX was positively correlated with expression of IL-1ß, tumor necrosis factor, and cyclooxygenase 2. Treatment with 17(R)-HDoHE reversed established pain behavior (but not joint pathology) in 2 models of OA pain. This was associated with a significant elevation in the plasma levels of resolvin D2 and a significant reduction in astrogliosis in the spinal cord in the monosodium iodoacetate-induced OA rat model. CONCLUSION: Our preclinical data demonstrate the robust analgesic effects of activation of the D series resolvin pathways in 2 different animal models of OA. Our data support a predominant central mechanism of action in clinically relevant models of OA pain.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Conducta Animal/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Osteoartritis de la Rodilla/genética , Receptores de Quimiocina/genética , Animales , Artralgia/inducido químicamente , Cartílago Articular/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/toxicidad , Expresión Génica , Humanos , Ácido Yodoacético/toxicidad , Meniscos Tibiales/cirugía , Neuroglía/citología , Neuroglía/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Quimiocina/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Receptores de Lipoxina/genética , Médula Espinal/citología , Médula Espinal/metabolismo , Membrana Sinovial/patología
3.
Physiol Rep ; 4(19)2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27694530

RESUMEN

Sjögren's syndrome (SS) is an autoimmune disorder characterized by chronic inflammation and destruction of salivary and lacrimal glands leading to dry mouth and dry eyes, respectively. Currently, the etiology of SS is unknown and the current therapies have no permanent benefit; therefore, new approaches are necessary to effectively treat this condition. Resolvins are highly potent endogenous lipid mediators that are synthesized during the resolution of inflammation to restore tissue homeostasis. Previous studies indicate that the resolvin family member, RvD1, binds to the ALX/FPR2 receptor to block inflammatory signals caused by tumor necrosis factor-alpha (TNF-α) in the salivary epithelium. More recently, the corticosteroid, dexamethasone (DEX), was shown to be effective in reducing salivary gland inflammation. However, DEX, as with other corticosteroids, elicits adverse secondary effects that could be ameliorated when used in smaller doses. Therefore, we investigated whether the more stable aspirin-triggered (AT) epimer, AT-RvD1, combined with reduced doses of DEX is effective in treating TNF-α-mediated disruption of polarized rat parotid gland (Par-C10) epithelial cell clusters. Our results indicate that AT-RvD1 and DEX individually reduced TNF-α-mediated alteration in the salivary epithelium (i.e, maintained cell cluster formation, increased lumen size, reduced apoptosis, and preserved cell survival signaling responses) as compared to untreated cells. Furthermore, AT-RvD1 combined with a reduced dose of DEX produced stronger responses (i.e., robust salivary cell cluster formation, larger lumen sizes, further reduced apoptosis, and sustained survival signaling responses) as compared to those observed with individual treatments. These studies demonstrate that AT-RvD1 combined with DEX is highly effective in treating TNF-α-mediated disruption of salivary gland epithelium.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Terapia Combinada/métodos , Dexametasona/farmacología , Ácidos Docosahexaenoicos/farmacología , Glucocorticoides/farmacología , Glándulas Salivales/citología , Síndrome de Sjögren/patología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Aspirina/administración & dosificación , Dexametasona/administración & dosificación , Ácidos Docosahexaenoicos/administración & dosificación , Glucocorticoides/administración & dosificación , Inflamación/metabolismo , Ratas , Receptores de Lipoxina/efectos de los fármacos , Receptores de Lipoxina/metabolismo , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/patología , Glándulas Salivales/ultraestructura , Transducción de Señal/fisiología , Síndrome de Sjögren/tratamiento farmacológico , Síndrome de Sjögren/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
4.
Stroke ; 47(2): 490-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26732571

RESUMEN

BACKGROUND AND PURPOSE: Lipoxin A4 (LXA4) has been reported to reduce inflammation in several neurological injury models. We studied the effects of LXA4 on neuroinflammation after subarachnoid hemorrhage (SAH) in a rat model. METHODS: Two hundred and thirty-eight Sprague-Dawley male rats, weight 280-320 g, were used. Exogenous LXA4 (0.3 and 1.0 nmol) were injected intracerebroventricularly at 1.5 hours after SAH. Neurological scores, brain water content, and blood-brain barrier were evaluated at 24 hours after SAH; Morris water maze and T-maze tests were examined at 21 days after SAH. The expression of endogenous LXA4 and its receptor formyl peptide receptor 2 (FPR2), as well as p38, interleukin-1ß, and interleukin-6 were studied either by ELISA or by Western blots. Neutrophil infiltration was observed by myeloperoxidase staining. FPR2 siRNA was used to knock down LXA4 receptor. RESULTS: The expression of endogenous LXA4 decreased, and the expression of FPR2 increased after SAH. Exogenous LXA4 decreased brain water content, reduced Evans blue extravasation, and improved neurological functions and improved the learning and memory ability after SAH. LXA4 reduced neutrophil infiltration and phosphorylation of p38, interleukin-1ß, and interleukin-6. These effects of LXA4 were abolished by FPR2 siRNA. CONCLUSIONS: Exogenous LXA4 inhibited inflammation by activating FPR2 and inhibiting p38 after SAH. LXA4 may serve as an alternative treatment to relieve early brain injury after SAH.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Conducta Animal/efectos de los fármacos , Lipoxinas/farmacología , Receptores de Lipoxina/efectos de los fármacos , Hemorragia Subaracnoidea/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Western Blotting , Edema Encefálico/inmunología , Edema Encefálico/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnicas de Silenciamiento del Gen , Inflamación , Inyecciones Intraventriculares , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Lipoxinas/inmunología , Lipoxinas/metabolismo , Masculino , Memoria/efectos de los fármacos , Pruebas Neuropsicológicas , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Ratas , Ratas Sprague-Dawley , Receptores de Lipoxina/genética , Receptores de Lipoxina/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Aprendizaje Espacial/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
J Leukoc Biol ; 97(4): 689-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25724390

RESUMEN

Most staphylococci produce short α-type PSMs and about twice as long ß-type PSMs that are potent leukocyte attractants and toxins. PSMs are usually secreted with the N-terminal formyl group but are only weak agonists for the leukocyte FPR1. Instead, the FPR1-related FPR2 senses PSMs efficiently and is crucial for leukocyte recruitment in infection. Which structural features distinguish FPR1 from FPR2 ligands has remained elusive. To analyze which peptide properties may govern the capacities of ß-type PSMs to activate FPRs, full-length and truncated variants of such peptides from Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis were synthesized. FPR2 activation was observed even for short N- or C-terminal ß-type PSM variants once they were longer than 18 aa, and this activity increased with length. In contrast, the shortest tested peptides were potent FPR1 agonists, and this property declined with increasing peptide length. Whereas full-length ß-type PSMs formed α-helices and exhibited no FPR1-specific activity, the truncated peptides had less-stable secondary structures, were weak agonists for FPR1, and required N-terminal formyl-methionine residues to be FPR2 agonists. Together, these data suggest that FPR1 and FPR2 have opposed ligand preferences. Short, flexible PSM structures may favor FPR1 but not FPR2 activation, whereas longer peptides with α-helical, amphipathic properties are strong FPR2 but only weak FPR1 agonists. These findings should help to unravel the ligand specificities of 2 critical human PRRs, and they may be important for new, anti-infective and anti-inflammatory strategies.


Asunto(s)
Proteínas Bacterianas/química , Toxinas Bacterianas/química , Proteínas Hemolisinas/química , Neutrófilos/efectos de los fármacos , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Secuencia de Aminoácidos , Proteínas Bacterianas/síntesis química , Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Antígeno CD11b/biosíntesis , Antígeno CD11b/genética , Señalización del Calcio/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Citotoxinas/síntesis química , Citotoxinas/química , Citotoxinas/farmacología , Células HL-60 , Proteínas Hemolisinas/síntesis química , Proteínas Hemolisinas/farmacología , Humanos , Interleucina-8/biosíntesis , Interleucina-8/genética , Datos de Secuencia Molecular , N-Formilmetionina/química , Neutrófilos/metabolismo , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Pliegue de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Staphylococcus aureus/química , Staphylococcus epidermidis/química , Staphylococcus lugdunensis/química , Relación Estructura-Actividad
6.
J Surg Res ; 183(2): 710-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23558258

RESUMEN

BACKGROUND: Hemorrhagic shock activates cellular stress signals and can lead to systemic inflammatory response, organ injury, and death. Mitogen-activated protein kinase (MAPK) acts as a sensor of tissue injury in models of ischemia-reperfusion injury. Lipoxins are endogenous lipid mediators with potent anti-inflammatory and pro-resolving actions. We hypothesized that BML-111 (a lipoxin A4-receptor agonist) attenuates hemorrhagic shock-induced acute lung injury (ALI) through inhibiting activation of the MAPK pathway. METHODS: We randomized Sprague-Dawley rats into four groups: sham, hemorrhagic shock-resuscitation (HS), HS plus BML-111 (BML-111), and HS plus BML-111 and BOC-2 (BOC-2). Two hours after resuscitation, we collected samples of lung. We obtained bronchoalveolar lavage fluid for neutrophil count. We performed optical microscopy to examine pathologic changes in lungs. Wet/dry ratios, myeloperoxidase expression, interleukin (IL)-1ß and IL-6 levels in lung were measured. We evaluated MAPK activation and the DNA binding activity of activator protein-1 in lung. RESULTS: Treatment with BML-111 reduced the lung damage and wet/dry ratio, neutrophil count in bronchoalveolar lavage fluid, expression of myeloperoxidase, and production of IL-1ß and IL-6 in lung. Phosphorylation of MAPK was also decreased by BML-111 in lung. Furthermore, the DNA binding activity of activator protein-1 was blocked by BML-111. An antagonist of the lipoxin A4-receptor, BOC-2, reversed the protective effect of BML-111 on ALI induced by hemorrhagic shock. CONCLUSIONS: This study indicates that BML-111 attenuated hemorrhagic shock-induced ALI via the MAPK/activator protein-1 signaling pathway. Therefore, BML-111 may have therapeutic potential for hemorrhagic shock-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/prevención & control , Ácidos Heptanoicos/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Choque Hemorrágico/complicaciones , Transducción de Señal/fisiología , Lesión Pulmonar Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Ácidos Heptanoicos/farmacología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Lipoxina/agonistas , Receptores de Lipoxina/antagonistas & inhibidores , Receptores de Lipoxina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/efectos de los fármacos , Factor de Transcripción AP-1/fisiología
7.
Arch Biochem Biophys ; 516(2): 121-7, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22005393

RESUMEN

Calpain inhibitors, including peptide aldehydes (N-acetyl-Leu-Leu-Nle-CHO and N-acetyl-Leu-Leu-Met-CHO) and α-mercapto-acrylic acid derivatives (PD150606 and PD151746), have been shown to stimulate phagocyte functions via activation of human formyl peptide receptor (hFPR) and/or hFPR-like 1 (hFPRL1). Using the homology modeling of the receptors and the ligand docking simulation, here we show that these calpain inhibitors could bind to the putative N-formyl-Met-Leu-Phe (fMLF) binding site on hFPR and/or hFPRL1. The studies with HEK-293 cells stably expressing hFPR or hFPRL1 showed that the concentrations of calpain inhibitors required to induce an increase in cytoplasmic free Ca(2+) ([Ca(2+)](i)) was much higher (>100 folds) than those of fMLF and Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm). HEK-293 cells expressing hFPR or hFPRL1 with the mutated fMLF binding site never exhibited the [Ca(2+)](i) response to calpain inhibitors. When the optimal concentrations of each stimulus were used, pretreatment of cells with fMLF or WKYMVm abolished an increase in [Ca(2+)](i) induced by calpain inhibitors as well as the same stimulus, whereas pretreatment of cells with calpain inhibitors significantly suppressed, but never abolished, the [Ca(2+)](i) response induced by fMLF or WKYMVm, suggesting that the binding affinity of the inhibitors to the putative fMLF binding site may be lower than that of fMLF or WKYMVm.


Asunto(s)
Glicoproteínas/farmacología , Receptores de Formil Péptido/química , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/química , Receptores de Lipoxina/efectos de los fármacos , Acrilatos/farmacología , Secuencia de Bases , Sitios de Unión , Señalización del Calcio/efectos de los fármacos , Simulación por Computador , Inhibidores de Cisteína Proteinasa/farmacología , Cartilla de ADN/genética , Células HEK293 , Humanos , Leupeptinas/farmacología , Ligandos , Modelos Moleculares , N-Formilmetionina Leucil-Fenilalanina/farmacología , Oligopéptidos/farmacología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/genética , Receptores de Lipoxina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología Estructural de Proteína
8.
ScientificWorldJournal ; 6: 1375-84, 2006 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-17072491

RESUMEN

The anti-inflammatory actions of the nonapeptide antiflammin-2, identified by homology with uteroglobin and annexin-A1 sequences, have been described in some detail, yet its mechanisms of action remain elusive. Since recent data indicate an involvement of the formyl peptide receptor (FPR)-like 1 (or FPRL-1) in the effects of annexin-A1, we have tested here the effect of antiflammin-2 with respect to this receptor family. Using HEK-293 cells expressing either human FPR and FPRL-1, and an annexin-A1 peptide as tracer ([125I-Tyr]-Ac2-26), we found that antiflammin-2 competed for binding only at FPRL-1, and not FPR, with an approximate EC50 of 1 mM. In line with data produced for the full-length protein, genuine receptor activation by antiflammin-2 was confirmed by rapid phosphorylation of extracellular-regulated kinase 1 and 2. Finally, study of the neutrophil interaction with activated endothelium under flow demonstrated an inhibitory effect of antiflammin-2, thus providing functional support to a role for the antiflammin-2/FPRL-1 anti-inflammatory axis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Unión Competitiva , Células Cultivadas , Humanos , Riñón/citología , Riñón/embriología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Receptores de Formil Péptido/fisiología , Receptores de Lipoxina/fisiología
9.
Inflamm Allergy Drug Targets ; 5(2): 107-14, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16613569

RESUMEN

This review will emphasize the concept of anti-inflammation and propose that better understanding of the resolution phase of the inflammatory response organized by the host against inflammatory insults can lead to the identification of novel targets for drug development. Under the umbrella of anti-inflammation, we discuss here recent discoveries in the biology of annexin 1 and glucocorticoids. The fact that annexin 1 and another anti-inflammatory mediator, lipoxin A4, converge onto a specific membrane receptor (termed ALX) might help understanding the resolution phase of inflammation, and strengthen the use of this target for innovative drug development. In addition, glucocorticoids (GC), historically linked to annexin 1, are widely use in the clinical control of several pathologies with an inflammatory etiology, though their use is often burdened by several side effects. The development of new GC with more specific or improved mechanisms, e.g. the nitro-steroids, would go along this wave and, potentially, could be of large applicability. The two mediators/targets here illustrated are to be taken as examples of the clues that the study of anti-inflammation might give to the pharmaceutical industry for innovative drug discovery.


Asunto(s)
Anexina A1/metabolismo , Antiinflamatorios/metabolismo , Glucocorticoides/metabolismo , Donantes de Óxido Nítrico/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Glucocorticoides/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipoxinas/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Prednisolona/análogos & derivados , Prednisolona/metabolismo , Prednisolona/farmacología , Prednisolona/uso terapéutico , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Receptores de Lipoxina/efectos de los fármacos
10.
Int J Immunopathol Pharmacol ; 17(2): 145-56, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15171815

RESUMEN

Lipoxin A4 (LXA4) is a potent eicosanoid that inhibits IL-1beta-induced activation of human fibroblast like synoviocytes (FLS) via the ALX4 receptor. Serum amyloid A (SAA) is an acute phase reactant with cytokine-like properties. SAA has been shown to bind the same seven transmembrane G protein-coupled receptor ligated by LXA4. Here we compared the inflammatory responses of lipid (LXA4) and peptide (SAA) ligands in human FLS via the shared ALX, and characterized their downstream signaling. LXA4 induced stimulation of tissue inhibitors of metalloproteinase-2, whereas SAA induced interleukin-8 and matrix metalloproteinase-3 production. SAA up-regulated NF-kappaB and AP-1 DNA binding activity, while LXA4 markedly inhibited these responses after IL-1beta stimulation. A human IL-8 promoter luciferase construct was transfected into CHO cells stably expressing ALXR in order to determine the role of NF-kappaB and/or AP-1 in the regulation of IL-8 gene expression. The NF-kappaB pathway proved to be the preeminent for the biological responses elicited by both ligands. These findings suggest that two endogenous molecules, targeting a common receptor, could participate in the pathogenesis of inflammatory arthritis by differentially regulating inflammatory responses in tissues expressing the ALXR.


Asunto(s)
Amiloide/farmacología , Interleucina-8/biosíntesis , Lipoxinas/farmacología , FN-kappa B/biosíntesis , Artritis Reumatoide/patología , Células Cultivadas , ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/metabolismo , Humanos , Interleucina-8/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , FN-kappa B/genética , Regiones Promotoras Genéticas/genética , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos , Inhibidores Tisulares de Metaloproteinasas/farmacología , Transfección
11.
J Immunol ; 172(11): 7078-85, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15153530

RESUMEN

Alzheimer's disease (AD) is characterized by overproduction of beta amyloid peptides in the brain with progressive loss of neuronal cells. The 42-aa form of the beta amyloid peptide (Abeta(42)) is implied as a major causative factor, because it is toxic to neurons and elicits inflammatory responses in the brain by activating microglial cells. Despite the overproduction of Abeta(42), AD brain tissue also generates protective factor(s) that may antagonize the neurodestructive effect of Abeta(42). Humanin is a gene cloned from an apparently normal region of an AD brain and encodes a 24-aa peptide. Both secreted and synthetic Humanin peptides protect neuronal cells from damage by Abeta(42), and the effect of Humanin may involve putative cellular receptor(s). To elucidate the molecular identity of such receptor(s), we examined the activity of synthetic Humanin on various cells and found that Humanin induced chemotaxis of mononuclear phagocytes by using a human G protein-coupled formylpeptide receptor-like-1 (FPRL1) and its murine counterpart FPR2. Coincidentally, FPRL1 and FPR2 are also functional receptors used by Abeta(42) to chemoattract and activate phagocytic cells. Humanin reduced the aggregation and fibrillary formation by suppressing the effect of Abeta(42) on mononuclear phagocytes. In neuroblast cells, Humanin and Abeta(42) both activated FPRL1; however, only Abeta(42) caused apoptotic death of the cells, and its cytopathic effect was blocked by Humanin. We conclude that Humanin shares human FPRL1 and mouse FPR2 with Abeta(42) and suggest that Humanin may exert its neuroprotective effects by competitively inhibiting the access of FPRL1 to Abeta(42).


Asunto(s)
Fármacos Neuroprotectores/farmacología , Proteínas/farmacología , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Enfermedad de Alzheimer/prevención & control , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Neuronas/efectos de los fármacos , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/agonistas
12.
Br J Pharmacol ; 141(1): 37-46, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14662730

RESUMEN

1. Human formyl peptide-receptor-like-1 (FPRL-1) is a promiscuous G protein-coupled receptor (GPCR), and belongs to a chemoattractant receptor family protein. This receptor has been reported to interact with various host-derived peptides and lipids involved in inflammatory responses. We described here, a novel role for FPRL-1 as a high-affinity beta-chemokine receptor for an N-terminally truncated form of the CKbeta8 (CCL23/MPIF-1) splice variant CKbeta8-1 (22-137 aa). 2. RT-PCR analysis of mRNA derived from human tissues and cells revealed a predominant expression of FPRL-1 in inflammatory cells, particularly in neutrophils. 3. Intracellular calcium mobilisation assay, used as screening tool, in recombinant Chinese hamster ovary (CHO-K1) and human embryonic kidney (HEK293s) cells coexpressing FPRL-1 and Galpha(16), demonstrated FPRL-1 is a functional high-affinity receptor for CKbeta8-1 (46-137 aa, sCKbeta8-1), with pEC(50) values of 9.13 and 8.85, respectively. 4. The FPRL-1 activation in CHO-K1 cells is mediated by Galpha(i)/Galpha(o) proteins, as assessed by pertussis toxin sensitivity and inhibition of forskolin-induced cyclic AMP accumulation. 5. Binding experiments were performed with a radio-iodinated synthetic peptide, [(125-)I]-WKYMVm, a known potent FPRL-1 agonist. CHO-K1 cell membranes expressing FPRL-1 bound [(125-)I]-WKYMVm with a K(d) value of 9.34. Many known FPRL-1 agonists were tested and sCKbeta8-1 was the most effective nonsynthetic ligand in displacing the radiolabelled agonist, with a pIC(50) of 7.97. 6. The functional significance of sCKbeta8-1 interaction with FPRL-1 was further demonstrated by the activation of polymorphonuclear leukocytes (PMNs) calcium mobilisation and chemotaxis. These interactions were shown to be via FPRL-1 by specific blockade of PMNs activation in the presence of an FPRL-1 antibody.


Asunto(s)
Quimiocinas CC/química , Quimiocinas CC/farmacología , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Lipoxina/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células CHO , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Quimiocinas CC/metabolismo , Quimiotaxis/efectos de los fármacos , Cricetinae , Evaluación Preclínica de Medicamentos/métodos , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Expresión Génica , Humanos , Radioisótopos de Yodo/metabolismo , Riñón/citología , Riñón/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/genética , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores Acoplados a Proteínas G , Receptores de Lipoxina/genética , Receptores de Lipoxina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...