Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 112022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-35293862

RESUMEN

Host immune responses contribute to dengue's pathogenesis and severity, yet the possibility that failure in endogenous inflammation resolution pathways could characterise the disease has not been contemplated. The pro-resolving protein Annexin A1 (AnxA1) is known to counterbalance overexuberant inflammation and mast cell (MC) activation. We hypothesised that inadequate AnxA1 engagement underlies the cytokine storm and vascular pathologies associated with dengue disease. Levels of AnxA1 were examined in the plasma of dengue patients and infected mice. Immunocompetent, interferon (alpha and beta) receptor one knockout (KO), AnxA1 KO, and formyl peptide receptor 2 (FPR2) KO mice were infected with dengue virus (DENV) and treated with the AnxA1 mimetic peptide Ac2-26 for analysis. In addition, the effect of Ac2-26 on DENV-induced MC degranulation was assessed in vitro and in vivo. We observed that circulating levels of AnxA1 were reduced in dengue patients and DENV-infected mice. Whilst the absence of AnxA1 or its receptor FPR2 aggravated illness in infected mice, treatment with AnxA1 agonistic peptide attenuated disease manifestationsatteanuated the symptoms of the disease. Both clinical outcomes were attributed to modulation of DENV-mediated viral load-independent MC degranulation. We have thereby identified that altered levels of the pro-resolving mediator AnxA1 are of pathological relevance in DENV infection, suggesting FPR2/ALX agonists as a therapeutic target for dengue disease.


Asunto(s)
Anexina A1 , Dengue , Animales , Anexina A1/metabolismo , Dengue/tratamiento farmacológico , Humanos , Inflamación/patología , Ratones , Péptidos/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo
2.
J Biol Chem ; 297(2): 100928, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34274316

RESUMEN

B-cell lymphoma 6 (BCL6) is a zinc finger transcriptional repressor possessing a BTB-POZ (BR-C, ttk, and bab for BTB; pox virus and zinc finger for POZ) domain, which is required for homodimerization and association with corepressors. BCL6 has multiple roles in normal immunity, autoimmunity, and some types of lymphoma. Mice bearing disrupted BCL6 loci demonstrate suppressed high-affinity antibody responses to T-dependent antigens. The corepressor binding groove in the BTB-POZ domain is a potential target for small compound-mediated therapy. Several inhibitors targeting this binding groove have been described, but these compounds have limited or absent in vivo activity. Biophysical studies of a novel compound, GSK137, showed an in vitro pIC50 of 8 and a cellular pIC50 of 7.3 for blocking binding of a peptide derived from the corepressor silencing mediator for retinoid or thyroid hormone receptors to the BCL6 BTB-POZ domain. The compound has good solubility (128 µg/ml) and permeability (86 nM/s). GSK137 caused little change in cell viability or proliferation in four BCL6-expressing B-cell lymphoma lines, although there was modest dose-dependent accumulation of G1 phase cells. Pharmacokinetic studies in mice showed a profile compatible with achieving good levels of target engagement. GSK137, administered orally, suppressed immunoglobulin G responses and reduced numbers of germinal centers and germinal center B cells following immunization of mice with the hapten trinitrophenol. Overall, we report a novel small-molecule BCL6 inhibitor with in vivo activity that inhibits the T-dependent antigen immune response.


Asunto(s)
Proteínas Proto-Oncogénicas c-bcl-6 , Animales , Linfocitos B/metabolismo , Humanos , Ratones , Transcripción Genética , Dedos de Zinc
3.
ILAR J ; 62(1-2): 66-76, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-35421235

RESUMEN

Animal studies in pharmaceutical drug discovery are common in preclinical research for compound evaluation before progression into human clinical trials. However, high rates of drug development attrition have prompted concerns regarding animal models and their predictive translatability to the clinic. To improve the characterization and evaluation of animal models for their translational relevance, the authors developed a tool to transparently reflect key features of a model that may be considered in both the application of the model but also the likelihood of successful translation of the outcomes to human patients. In this publication, we describe the rationale for the development of the Animal Model Quality Assessment tool, the questions used for the animal model assessment, and a high-level scoring system for the purpose of defining predictive translatability. Finally, we provide an example of a completed Animal Model Quality Assessment for the adoptive T-cell transfer model of colitis as a mouse model to mimic inflammatory bowel disease in humans.


Asunto(s)
Modelos Animales de Enfermedad , Descubrimiento de Drogas , Animales , Humanos , Ratones
5.
Science ; 368(6489): 387-394, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32193360

RESUMEN

The two tandem bromodomains of the BET (bromodomain and extraterminal domain) proteins enable chromatin binding to facilitate transcription. Drugs that inhibit both bromodomains equally have shown efficacy in certain malignant and inflammatory conditions. To explore the individual functional contributions of the first (BD1) and second (BD2) bromodomains in biology and therapy, we developed selective BD1 and BD2 inhibitors. We found that steady-state gene expression primarily requires BD1, whereas the rapid increase of gene expression induced by inflammatory stimuli requires both BD1 and BD2 of all BET proteins. BD1 inhibitors phenocopied the effects of pan-BET inhibitors in cancer models, whereas BD2 inhibitors were predominantly effective in models of inflammatory and autoimmune disease. These insights into the differential requirement of BD1 and BD2 for the maintenance and induction of gene expression may guide future BET-targeted therapies.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Histona Acetiltransferasas/antagonistas & inhibidores , Factores Inmunológicos/farmacología , Terapia Molecular Dirigida , Factores de Transcripción/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Histona Acetiltransferasas/química , Histona Acetiltransferasas/genética , Humanos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Factores Inmunológicos/química , Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Dominios Proteicos/efectos de los fármacos , Factores de Transcripción/química , Factores de Transcripción/genética
6.
J Clin Invest ; 130(3): 1156-1167, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32015229

RESUMEN

Understanding the circuits that promote an efficient resolution of inflammation is crucial to deciphering the molecular and cellular processes required to promote tissue repair. Macrophages play a central role in the regulation of inflammation, resolution, and repair/regeneration. Using a model of skeletal muscle injury and repair, herein we identified annexin A1 (AnxA1) as the extracellular trigger of macrophage skewing toward a pro-reparative phenotype. Brought into the injured tissue initially by migrated neutrophils, and then overexpressed in infiltrating macrophages, AnxA1 activated FPR2/ALX receptors and the downstream AMPK signaling cascade, leading to macrophage skewing, dampening of inflammation, and regeneration of muscle fibers. Mice lacking AnxA1 in all cells or only in myeloid cells displayed a defect in this reparative process. In vitro experiments recapitulated these properties, with AMPK-null macrophages lacking AnxA1-mediated polarization. Collectively, these data identified the AnxA1/FPR2/AMPK axis as an important pathway in skeletal muscle injury regeneration.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Anexina A1/metabolismo , Músculo Esquelético , Regeneración , Transducción de Señal , Proteínas Quinasas Activadas por AMP/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Anexina A1/genética , Ratones , Ratones Noqueados , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo
7.
Res Vet Sci ; 125: 434-440, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29108655

RESUMEN

Proteinase-activated receptor 2 (PAR2) is a G-protein-coupled receptor for trypsin and mast cell tryptase; it is highly expressed at the intestinal level with multiple functions, such as epithelial permeability and intestinal motility. The aim of the study was to evaluate the distribution and expression of proteinase-activated receptor 2 in the small intestine during herniation through epiploic foramen. In this prospective clinical study, eight horses admitted for colic and which underwent exploratory laparotomy were considered. During surgery, the jejunum or the ileum was sampled by enterectomy. Morphological examination (histology, PAR2 immunohistochemistry) and molecular biology analysis (western blot and quantitative polymerase chain reaction) were carried out on the resected intestinal samples. The Marginal Injured Tracts (MITs) and Central Injury Tracts (CITs) were defined as the oral and caudal marginal segments of the resected bowel tract and as the geometric centre of the intestinal ischaemic lesion length, respectively. The PAR2 immunoreactivity was particularly evident in the epithelial cells, with higher immunoreactivity in the MIT rather than in the CIT. Moreover, a different immune localisation was observed in the MITs at the cell membrane level and in the CITs in the cytoplasm. No statistical difference was observed in PAR2 mRNA and protein (44kDa) expression between the MIT and the CIT. The PAR2 protein content in the intestinal tracts which were removed from horses with herniation was lower when compared with the control animals. This study provided data concerning the PAR2 presence and distribution in horses with intestinal herniation through the epiploic foramen.


Asunto(s)
Enfermedades de los Caballos/metabolismo , Intestino Delgado/patología , Receptor PAR-2/metabolismo , Animales , Cólico/patología , Regulación de la Expresión Génica , Caballos , Íleon/metabolismo , Laparotomía , Estudios Prospectivos , Receptor PAR-2/genética , Tripsina/metabolismo
8.
JCI Insight ; 2(7): e90723, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28405616

RESUMEN

Living in a mentally and physically stimulating environment has been suggested to have a beneficial effect on the immune response. This study investigates these effects, utilizing a 2-week program of environmental enrichment (EE) and 2 models of acute inflammation: zymosan-induced peritonitis (ZIP) and the cecal ligation and puncture (CLP) model of sepsis. Our results revealed that following exposure to EE, mice possessed a significantly higher circulating neutrophil to lymphocyte ratio compared with control animals. When subject to ZIP, EE animals exhibit enhanced neutrophil and macrophage influx into their peritoneal cavity. Corresponding results were found in CLP, where we observed an improved capacity for enriched animals to clear systemic microbial infection. Ex vivo investigation of leukocyte activity also revealed that macrophages from EE mice presented an enhanced phagocytic capacity. Supporting these findings, microarray analysis of EE animals revealed the increased expression of immunomodulatory genes associated with a heightened and immunoprotective status. Taken together, these results provide potentially novel mechanisms by which EE influences the development and dynamics of the immune response.


Asunto(s)
Ambiente , Inflamación/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Peritonitis/inmunología , Sepsis/inmunología , Animales , Ciego/lesiones , Modelos Animales de Enfermedad , Ligadura , Masculino , Ratones , Análisis por Micromatrices , Peritonitis/inducido químicamente , Fagocitosis , Sepsis/etiología , Zimosan
9.
Proc Natl Acad Sci U S A ; 114(15): 3963-3968, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28356517

RESUMEN

The resolution of inflammation is an active process orchestrated by specialized proresolving lipid mediators (SPM) that limit the host response within the affected tissue; failure of effective resolution may lead to tissue injury. Because persistence of inflammatory signals is a main feature of chronic inflammatory conditions, including inflammatory bowel diseases (IBDs), herein we investigate expression and functions of SPM in intestinal inflammation. Targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics was used to identify SPMs from n-3 polyunsaturated fatty acids in human IBD colon biopsies, quantifying a significant up-regulation of the resolvin and protectin pathway compared with normal gut tissue. Systemic treatment with protectin (PD)1n-3 DPA or resolvin (Rv)D5n-3 DPA protected against colitis and intestinal ischemia/reperfusion-induced inflammation in mice. Inhibition of 15-lipoxygenase activity reduced PD1n-3 DPA and augmented intestinal inflammation in experimental colitis. Intravital microscopy of mouse mesenteric venules demonstrated that PD1n-3 DPA and RvD5n-3 DPA decreased the extent of leukocyte adhesion and emigration following ischemia-reperfusion. These data were translated by assessing human neutrophil-endothelial interactions under flow: PD1n-3 DPA and RvD5n-3 DPA reduced cell adhesion onto TNF-α-activated human endothelial monolayers. In conclusion, we propose that innovative therapies based on n-3 DPA-derived mediators could be developed to enable antiinflammatory and tissue protective effects in inflammatory pathologies of the gut.


Asunto(s)
Colitis/prevención & control , Ácidos Docosahexaenoicos/farmacología , Enfermedades Inflamatorias del Intestino/metabolismo , Intestinos/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Colitis/inducido químicamente , Ácidos Docosahexaenoicos/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Mesenterio/irrigación sanguínea , Mesenterio/efectos de los fármacos , Ratones Endogámicos C57BL , Persona de Mediana Edad , Peritonitis/inducido químicamente , Peritonitis/prevención & control , Daño por Reperfusión/prevención & control
10.
J Am Soc Nephrol ; 28(1): 94-105, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27153924

RESUMEN

Patients with CKD requiring dialysis have a higher risk of sepsis and a 100-fold higher mortality rate than the general population with sepsis. The severity of cardiac dysfunction predicts mortality in patients with sepsis. Here, we investigated the effect of preexisting CKD on cardiac function in mice with sepsis and whether inhibition of IκB kinase (IKK) reduces the cardiac dysfunction in CKD sepsis. Male C57BL/6 mice underwent 5/6 nephrectomy, and 8 weeks later, they were subjected to LPS (2 mg/kg) or sepsis by cecal ligation and puncture (CLP). Compared with sham operation, nephrectomy resulted in significant increases in urea and creatinine levels, a small (P<0.05) reduction in ejection fraction (echocardiography), and increases in the cardiac levels of phosphorylated IκBα, Akt, and extracellular signal-regulated kinase 1/2; nuclear translocation of the NF-κB subunit p65; and inducible nitric oxide synthase (iNOS) expression. When subjected to LPS or CLP, compared with sham-operated controls, CKD mice exhibited exacerbation of cardiac dysfunction and lung inflammation, greater increases in levels of plasma cytokines (TNF-α, IL-1ß, IL-6, and IL-10), and greater increases in the cardiac levels of phosphorylated IKKα/ß and IκBα, nuclear translocation of p65, and iNOS expression. Treatment of CKD mice with an IKK inhibitor (IKK 16; 1 mg/kg) 1 hour after CLP or LPS administration attenuated these effects. Thus, preexisting CKD aggravates the cardiac dysfunction caused by sepsis or endotoxemia in mice; this effect may be caused by increased cardiac NF-κB activation and iNOS expression.


Asunto(s)
Cardiopatías/enzimología , Cardiopatías/prevención & control , Quinasa I-kappa B/antagonistas & inhibidores , Insuficiencia Renal Crónica/enzimología , Sepsis/complicaciones , Animales , Cardiopatías/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/complicaciones
11.
Biol Chem ; 397(10): 981-93, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27447237

RESUMEN

Inflammation is essential to protect the host from exogenous and endogenous dangers that ultimately lead to tissue injury. The consequent tissue repair is intimately associated with the fate of the inflammatory response. Restoration of tissue homeostasis is achieved through a balance between pro-inflammatory and anti-inflammatory/pro-resolving mediators. In chronic inflammatory diseases such balance is compromised, resulting in persistent inflammation and impaired healing. During the last two decades the glucocorticoid-regulated protein Annexin A1 (AnxA1) has emerged as a potent pro-resolving mediator acting on several facets of the innate immune system. Here, we review the therapeutic effects of AnxA1 on tissue healing and repairing together with the molecular targets responsible for these complex biological properties.


Asunto(s)
Anexina A1/metabolismo , Transducción de Señal , Animales , Humanos , Inflamación/metabolismo , Inflamación/patología
12.
Br J Pharmacol ; 173(9): 1556-65, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26890936

RESUMEN

BACKGROUND AND PURPOSE: Hydrogen sulfide (H2S) is a gasotransmitter produced from L-cysteine through the enzymatic action of cystathionine-γ-lyase (CSE) and/or cystathionine-ß-synthase. D-Penicillamine is the d isomer of a dimethylated cysteine and has been used for the treatment of rheumatoid arthritis. AsD-penicillamine is structurally very similar to cysteine, we have investigated whether D-penicillamine, as a cysteine analogue, has an effect on the H2 S pathway. EXPERIMENTAL APPROACH: We tested the effect of D-penicillamine (0.01-1 mM) in mouse aortic rings mounted in isolated organ baths and determined whether it could affect H2 S biosynthesis. In particular, we investigated any possible inhibitor or donor behaviour by using recombinant enzyme-based assays and an in vivo approach. KEY RESULTS: D-Penicillamine, per se, showed little or no vasodilator effect, and it cannot be metabolized as a substrate in place of l-cysteine. However, d-penicillamine significantly reduced L-cysteine-induced vasodilatation in a concentration-dependent manner through inhibition of H2 S biosynthesis, and this effect occurred at concentrations 10 times lower than those needed to induce the release of H2 S. In particular, D-penicillamine selectively inhibited CSE in a pyridoxal-5'-phosphate-dependent manner. CONCLUSIONS AND IMPLICATIONS: Taken together, our results suggest that D-penicillamine acts as a selective CSE inhibitor, leading to new perspectives in the design and use of specific pharmacological tools for H2 S research. In addition, the inhibitory effect of D-penicillamine on CSE could account for its beneficial action in rheumatoid arthritis patients, where H2 S has been shown to have a detrimental effect.


Asunto(s)
Cistationina gamma-Liasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Sulfuro de Hidrógeno/metabolismo , Penicilamina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Cistationina gamma-Liasa/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Masculino , Ratones , Penicilamina/química , Relación Estructura-Actividad
13.
J Control Release ; 217: 284-92, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26385167

RESUMEN

Synthetic microstructures can be engineered to deliver bioactive compounds impacting on their pharmacokinetics and pharmacodynamics. Herein, we applied dextran-based layer-by-layer (LbL) microcapsules to deliver alpha-2-macroglobulin (α2MG), a protein with modulatory properties in inflammation. Extending recent observations made with dextran-microcapsules loaded with α2MG in experimental sepsis, we focused on the physical and chemical characteristics of these microstructures and determined their biology on rodent and human cells. We report an efficient encapsulation of α2MG into microcapsules, which enhanced i) human leukocyte recruitment to inflamed endothelium and ii) human macrophage phagocytosis: in both settings microcapsules were more effective than soluble α2MG or empty microcapsules (devoid of active protein). Translation of these findings revealed that intravenous administration of α2MG-microcapsules (but not empty microcapsules) promoted neutrophil migration into peritoneal exudates and augmented macrophage phagocytic functions, the latter response being associated with alteration of bioactive lipid mediators as assessed by mass spectrometry. The present study indicates that microencapsulation can be an effective strategy to harness the complex biology of α2MG with enhancing outcomes on fundamental processes of the innate immune response paving the way to potential future development in the control of sepsis.


Asunto(s)
Leucocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , alfa-Macroglobulinas/administración & dosificación , Animales , Cápsulas , Células Cultivadas , Dextranos/química , Composición de Medicamentos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunidad Innata/efectos de los fármacos , Leucocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , alfa-Macroglobulinas/farmacología
14.
J Immunol ; 195(3): 1139-51, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26101324

RESUMEN

Blood-derived monocytes remove apoptotic cells and terminate inflammation in settings as diverse as atherosclerosis and Alzheimer's disease. They express high levels of the proresolving receptor ALX/FPR2, which is activated by the protein annexin A1 (ANXA1), found in high abundance in inflammatory exudates. Using primary human blood monocytes from healthy donors, we identified ANXA1 as a potent CD14(+)CD16(-) monocyte chemoattractant, acting via ALX/FPR2. Downstream signaling pathway analysis revealed the p38 MAPK-mediated activation of a calcium independent phospholipase A2 with resultant synthesis of lysophosphatidic acid (LPA) driving chemotaxis through LPA receptor 2 and actin cytoskeletal mobilization. In vivo experiments confirmed ANXA1 as an independent phospholipase A2-dependent monocyte recruiter; congruently, monocyte recruitment was significantly impaired during ongoing zymosan-induced inflammation in AnxA1(-/-) or alx/fpr2/3(-/-) mice. Using a dorsal air-pouch model, passive transfer of apoptotic neutrophils between AnxA1(-/-) and wild-type mice identified effete neutrophils as the primary source of soluble ANXA1 in inflammatory resolution. Together, these data elucidate a novel proresolving network centered on ANXA1 and LPA generation and identify previously unappreciated determinants of ANXA1 and ALX/FPR2 signaling in monocytes.


Asunto(s)
Anexina A1/inmunología , Apoptosis/inmunología , Monocitos/inmunología , Neutrófilos/inmunología , Receptores del Ácido Lisofosfatídico/inmunología , Citoesqueleto de Actina/metabolismo , Animales , Anexina A1/genética , Células Cultivadas , Activación Enzimática/inmunología , Humanos , Inflamación/inmunología , Receptores de Lipopolisacáridos/metabolismo , Lisofosfolípidos/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/trasplante , Fosfolipasas A2 Calcio-Independiente/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Formil Péptido/biosíntesis , Receptores de Formil Péptido/genética , Receptores de Formil Péptido/metabolismo , Receptores de IgG/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Zimosan , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Immunol ; 194(7): 3381-8, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25725103

RESUMEN

There is a need for novel approaches to control pathologies with overexuberant inflammatory reactions. Targeting melanocortin (MC) receptors represents a promising therapy for obesity and chronic inflammation, but lack of selectivity and safety concerns limit development. A new way to increase selectivity of biological effects entails the identification of biased agonists. In this study, we characterize the small molecule AP1189 as a biased agonist at receptors MC1 and MC3. Although not provoking canonical cAMP generation, AP1189 addition to MC1 or MC3, but not empty vector, transfected HEK293 cells caused ERK1/2 phosphorylation, a signaling responsible for the proefferocytic effect evoked in mouse primary macrophages. Added to macrophage cultures, AP1189 reduced cytokine release, an effect reliant on both MC1 and MC3 as evident from the use of Mc1r(-/-) and Mc3r(-/-) macrophages. No melanogenesis was induced by AP1189 in B16-F10 melanocytes. In vivo, oral AP1189 elicited anti-inflammatory actions in peritonitis and, upon administration at the peak of inflammation, accelerated the resolution phase by ∼3-fold. Finally, given the clinical efficacy of adrenocorticotropin in joint diseases, AP1189 was tested in experimental inflammatory arthritis, where this biased agonist afforded significant reduction of macroscopic and histological parameters of joint disruption. These proof-of-concept analyses with AP1189, an active oral anti-inflammatory and resolution-promoting compound, indicate that biased agonism at MC receptors is an innovative, viable approach to yield novel anti-inflammatory molecules endowed with a more favorable safety profile.


Asunto(s)
Guanidinas/farmacología , Pirroles/farmacología , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/metabolismo , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/genética , Artritis Experimental/metabolismo , Artritis Experimental/patología , Calcio/metabolismo , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Guanidinas/administración & dosificación , Células HEK293 , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Melaninas/metabolismo , Melanoma Experimental , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Peritonitis/genética , Peritonitis/metabolismo , Peritonitis/patología , Fagocitosis/inmunología , Pirroles/administración & dosificación , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Receptores de Melanocortina/genética , Transducción de Señal/efectos de los fármacos
16.
Proc Natl Acad Sci U S A ; 111(52): 18685-90, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512512

RESUMEN

Sepsis is characterized by overlapping phases of excessive inflammation temporally aligned with an immunosuppressed state, defining a complex clinical scenario that explains the lack of successful therapeutic options. Here we tested whether the formyl-peptide receptor 2/3 (Fpr2/3)--ortholog to human FPR2/ALX (receptor for lipoxin A4)--exerted regulatory and organ-protective functions in experimental sepsis. Coecal ligature and puncture was performed to obtain nonlethal polymicrobial sepsis, with animals receiving antibiotics and analgesics. Clinical symptoms, temperature, and heart function were monitored up to 24 h. Peritoneal lavage and plasma samples were analyzed for proinflammatory and proresolving markers of inflammation and organ dysfunction. Compared with wild-type mice, Fpr2/3(-/-) animals exhibited exacerbation of disease severity, including hypothermia and cardiac dysfunction. This scenario was paralleled by higher levels of cytokines [CXCL1 (CXC receptor ligand 1), CCL2 (CC receptor ligand 2), and TNFα] as quantified in cell-free biological fluids. Reduced monocyte recruitment in peritoneal lavages of Fpr2/3(-/-) animals was reflected by a higher granulocyte/monocyte ratio. Monitoring Fpr2/3(-/-) gene promoter activity with a GFP proxy marker revealed an over threefold increase in granulocyte and monocyte signals at 24 h post-coecal ligature and puncture, a response mediated by TNFα. Treatment with a receptor peptido-agonist conferred protection against myocardial dysfunction in wild-type, but not Fpr2/3(-/-), animals. Therefore, coordinated physio-pharmacological analyses indicate nonredundant modulatory functions for Fpr2/3 in experimental sepsis, opening new opportunities to manipulate the host response for therapeutic development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Granulocitos/metabolismo , Monocitos/metabolismo , Receptores de Formil Péptido/metabolismo , Sepsis/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Modelos Animales de Enfermedad , Granulocitos/patología , Humanos , Ratones , Ratones Noqueados , Monocitos/patología , Peritoneo/metabolismo , Peritoneo/patología , Receptores de Formil Péptido/genética , Sepsis/genética , Sepsis/patología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
PLoS One ; 9(12): e114626, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25517119

RESUMEN

Formyl peptide receptors (FPR) belong to a family of sensors of the immune system that detect microbe-associated molecules and inform various cellular and sensorial mechanisms to the presence of pathogens in the host. Here we demonstrate that Fpr2/3-deficient mice show a distinct profile of behaviour characterised by reduced anxiety in the marble burying and light-dark box paradigms, increased exploratory behaviour in an open-field, together with superior performance on a novel object recognition test. Pharmacological blockade with a formyl peptide receptor antagonist, Boc2, in wild type mice reproduced most of the behavioural changes observed in the Fpr2/3(-/-) mice, including a significant improvement in novel object discrimination and reduced anxiety in a light/dark shuttle test. These effects were associated with reduced FPR signalling in the gut as shown by the significant reduction in the levels of p-p38. Collectively, these findings suggest that homeostatic FPR signalling exerts a modulatory effect on anxiety-like behaviours. These findings thus suggest that therapies targeting FPRs may be a novel approach to ameliorate behavioural abnormalities present in neuropsychiatric disorders at the cognitive-emotional interface.


Asunto(s)
Ansiedad/complicaciones , Ansiedad/tratamiento farmacológico , Terapia Molecular Dirigida , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Receptores de Formil Péptido/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/patología , Corticosterona/sangre , Conducta Exploratoria/efectos de los fármacos , Eliminación de Gen , Masculino , Ratones , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/deficiencia , Receptores de Formil Péptido/genética , Reconocimiento en Psicología/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
18.
Proc Natl Acad Sci U S A ; 110(45): 18232-7, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24108355

RESUMEN

Formyl-peptide receptor type 2 (FPR2), also called ALX (the lipoxin A4 receptor), conveys the proresolving properties of lipoxin A4 and annexin A1 (AnxA1) and the proinflammatory signals elicited by serum amyloid protein A and cathelicidins, among others. We tested here the hypothesis that ALX might exist as homo- or heterodimer with FPR1 or FPR3 (the two other family members) and operate in a ligand-biased fashion. Coimmunoprecipitation and bioluminescence resonance energy transfer assays with transfected HEK293 cells revealed constitutive dimerization of the receptors; significantly, AnxA1, but not serum amyloid protein A, could activate ALX homodimers. A p38/MAPK-activated protein kinase/heat shock protein 27 signaling signature was unveiled after AnxA1 application, leading to generation of IL-10, as measured in vitro (in primary monocytes) and in vivo (after i.p. injection in the mouse). The latter response was absent in mice lacking the ALX ortholog. Using a similar approach, ALX/FPR1 heterodimerization evoked using the panagonist peptide Ac2-26, identified a JNK-mediated proapoptotic path that was confirmed in primary neutrophils. These findings provide a molecular mechanism that accounts for the dual nature of ALX and indicate that agonist binding and dimerization state contribute to the conformational landscape of FPRs.


Asunto(s)
Anexina A1/metabolismo , Conformación Proteica , Receptores de Formil Péptido/química , Receptores de Formil Péptido/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Transferencia de Energía por Resonancia de Bioluminiscencia , Dimerización , Células HEK293 , Humanos , Inmunoprecipitación , Interleucina-10/metabolismo , Ratones , Datos de Secuencia Molecular , Proteína Amiloide A Sérica/metabolismo
19.
PLoS One ; 8(9): e75581, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24073272

RESUMEN

Polyunsaturated fatty acid (PUFA) metabolites are bioactive autoacoids that play an important role in the pathogenesis of a vast number of pathologies, including gut diseases. The induction and the resolution of inflammation depend on PUFA metabolic pathways that are favored. Therefore, understanding the profile of n-6 (eicosanoids)/n-3 (docosanoids) PUFA-derived metabolites appear to be as important as gene or protein array approaches, to uncover the molecules potentially implicated in inflammatory diseases. Using high sensitivity liquid chromatography tandem mass spectrometry, we characterized the tissue profile of PUFA metabolites in an experimental model of murine intestinal ischemia reperfusion. We identified temporal and quantitative differences in PUFA metabolite production, which correlated with inflammatory damage. Analysis revealed that early ischemia induces both pro-inflammatory and anti-inflammatory eicosanoid production. Primarily, LOX- (5/15/12/8-HETE, LTB4, LxA4) and CYP- (5, 6-EET) metabolites were produced upon ischemia, but also PGE3, and PDx. This suggests that different lipids simultaneously play a role in the induction and counterbalance of ischemic inflammatory response from its onset. COX-derived metabolites were more present from 2 to 5 hours after reperfusion, fitting with the concomitant inflammatory peaks. All metabolites were decreased 48 hours post-reperfusion except for to the pro-resolving RvE precursor 18-HEPE and the PPAR-Î³αµµα agonist, 15d-PGJ2. Data obtained through the pharmacological blockade of transient receptor potential vanilloid-4, which can be activated by 5, 6-EET, revealed that the endogenous activation of this receptor modulates post-ischemic intestinal inflammation. Altogether, these results demonstrate that different lipid pathways are involved in intestinal ischemia-reperfusion processes. Some metabolites, which expression is severely changed upon intestinal ischemia-reperfusion could provide novel targets and may facilitate the development of new pharmacological treatments.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Inflamación/metabolismo , Intestino Delgado/metabolismo , Isquemia/metabolismo , Daño por Reperfusión/metabolismo , Animales , Cromatografía Liquida , Citocinas/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Intestino Delgado/patología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Daño por Reperfusión/patología , Tasa de Supervivencia , Espectrometría de Masas en Tándem
20.
Blood ; 122(4): 608-17, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23733341

RESUMEN

Endogenous protective pathways mitigate the overshooting of inflammation after sterile or infectious injury. Here we report that formyl peptide receptor 2 (Fpr2/3) null mice display a major phenotype with exacerbated vascular inflammation observed postischemia reperfusion (IR) injury of the mesenteric artery, characterized by marked neutrophil adhesion and extravasation as visualized by intravital microscopy. Analysis of endogenous agonists for Fpr2/3 revealed that lipoxin A4 (LXA4) was generated by platelet/neutrophil aggregates during ischemia: this cellular response was attenuated in Fpr2/3(-/-) mice; hence, LXA4 levels were lower after 30 minutes' ischemia, and associated with augmented vascular inflammation in the reperfusion (45-180 minutes) phase. Exogenous delivery of LXA4 attenuated IR-mediated inflammation in Fpr2/3(+/+) but not Fpr2/3(-/-) mice; conversely, an Fpr2/3 antagonist skewed the vascular phenotype of Fpr2/3(+/+) mice to that of Fpr2/3(-/-) animals. Such LXA4-based circuit could be activated by aspirin (30-100 mg/kg), which triggered formation of 15-epi-LXA4 in wild-type mice, yet it was effective in Fpr2/3(-/-) mice. In summary, we propose that during ischemia, neutrophil Fpr2/3 controls platelet/neutrophil aggregates with the rapid generation of circulating LXA4, which in turn modulates downstream vascular inflammatory responses evident during the reperfusion phase.


Asunto(s)
Aspirina/farmacología , Vasos Sanguíneos , Citoprotección , Lipoxinas/farmacología , Microcirculación/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Citoprotección/efectos de los fármacos , Citoprotección/genética , Lipoxinas/química , Masculino , Ratones , Ratones Noqueados , Microcirculación/genética , Modelos Biológicos , Profármacos/química , Profármacos/farmacología , Receptores de Formil Péptido/genética , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...