Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(21)2023 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-37958978

RESUMEN

Neuroinflammation driven primarily by microglia directly contributes to neuronal death in many neurodegenerative diseases. Classical anti-inflammatory approaches aim to suppress pro-inflammatory mediator production, but exploitation of inflammatory resolution may also be of benefit. A key driver of peripheral inflammatory resolution, formyl peptide receptor 2 (Fpr2), is expressed by microglia, but its therapeutic potential in neurodegeneration remains unclear. Here, we studied whether targeting of Fpr2 could reverse inflammatory microglial activation induced by the potent bacterial inflammogen lipopolysaccharide (LPS). Exposure of murine primary or immortalised BV2 microglia to LPS triggered pro-inflammatory phenotypic change and activation of ROS production, effects significantly attenuated by subsequent treatment with the Fpr2 agonist C43. Mechanistic studies showed C43 to act through p38 MAPK phosphorylation and reduction of LPS-induced NFκB nuclear translocation via prevention of IκBα degradation. Here, we provide proof-of-concept data highlighting Fpr2 as a potential target for control of microglial pro-inflammatory activity, suggesting that it may be a promising therapeutic target for the treatment of neuroinflammatory disease.


Asunto(s)
Lipopolisacáridos , Microglía , Animales , Ratones , Antiinflamatorios/farmacología , Muerte Celular , Lipopolisacáridos/farmacología , Microglía/metabolismo , FN-kappa B/metabolismo
2.
FEBS J ; 289(7): 1801-1822, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33811735

RESUMEN

Alzheimer's disease and dementia are among the most significant current healthcare challenges given the rapidly growing elderly population, and the almost total lack of effective therapeutic interventions. Alzheimer's disease pathology has long been considered in terms of accumulation of amyloid beta and hyperphosphorylated tau, but the importance of neuroinflammation in driving disease has taken greater precedence over the last 15-20 years. Inflammatory activation of the primary brain immune cells, the microglia, has been implicated in Alzheimer's pathogenesis through genetic, preclinical, imaging and postmortem human studies, and strategies to regulate microglial activity may hold great promise for disease modification. Neuroinflammation is necessary for defence of the brain against pathogen invasion or damage but is normally self-limiting due to the engagement of endogenous pro-resolving circuitry that terminates inflammatory activity, a process that appears to fail in Alzheimer's disease. Here, we discuss the potential for a major regulator and promoter of resolution, the receptor FPR2, to restrain pro-inflammatory microglial activity, and propose that it may serve as a valuable target for therapeutic investigation in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Humanos , Microglía/metabolismo , Receptores de Formil Péptido/genética
3.
Cells ; 11(1)2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-35011694

RESUMEN

Increased osteoclast (OC) differentiation and activity is the critical event that results in bone loss and joint destruction in common pathological bone conditions, such as osteoporosis and rheumatoid arthritis (RA). RANKL and its decoy receptor, osteoprotegerin (OPG), control OC differentiation and activity. However, there is a specific concern of a rebound effect of denosumab discontinuation in treating osteoporosis. TNFα can induce OC differentiation that is independent of the RANKL/RANK system. In this review, we discuss the factors that negatively and positively regulate TNFα induction of OC formation, and the mechanisms involved to inform the design of new anti-resorptive agents for the treatment of bone conditions with enhanced OC formation. Similar to, and being independent of, RANKL, TNFα recruits TNF receptor-associated factors (TRAFs) to sequentially activate transcriptional factors NF-κB p50 and p52, followed by c-Fos, and then NFATc1 to induce OC differentiation. However, induction of OC formation by TNFα alone is very limited, since it also induces many inhibitory proteins, such as TRAF3, p100, IRF8, and RBP-j. TNFα induction of OC differentiation is, however, versatile, and Interleukin-1 or TGFß1 can enhance TNFα-induced OC formation through a mechanism which is independent of RANKL, TRAF6, and/or NF-κB. However, TNFα polarized macrophages also produce anabolic factors, including insulin such as 6 peptide and Jagged1, to slow down bone loss in the pathological conditions. Thus, the development of novel approaches targeting TNFα signaling should focus on its downstream molecules that do not affect its anabolic effect.


Asunto(s)
Diferenciación Celular , Osteoclastos/citología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Artritis Reumatoide/complicaciones , Diferenciación Celular/efectos de los fármacos , Humanos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoporosis/complicaciones , Transducción de Señal/efectos de los fármacos
4.
Oxid Med Cell Longev ; 2020: 2139192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32617132

RESUMEN

Microglial inflammatory activity is thought to be a major contributor to the pathology of neurodegenerative conditions such as Alzheimer's disease (AD), and strategies to restrain their behaviour are under active investigation. Classically, anti-inflammatory approaches are aimed at suppressing proinflammatory mediator production, but exploitation of inflammatory resolution, the endogenous process whereby an inflammatory reaction is terminated, has not been fully investigated as a therapeutic approach in AD. In this study, we sought to provide proof-of-principle that the major proresolving actor, formyl peptide receptor 2, Fpr2, could be targeted to reverse microglial activation induced by the AD-associated proinflammatory stimulus, oligomeric ß-amyloid (oAß). The immortalised murine microglial cell line BV2 was employed as a model system to investigate the proresolving effects of the Fpr2 ligand QC1 upon oAß-induced inflammatory, oxidative, and metabolic behaviour. Cytotoxic behaviour of BV2 cells was assessed through the use of cocultures with retinoic acid-differentiated human SH-SY5Y cells. Stimulation of BV2 cells with oAß at 100 nM did not induce classical inflammatory marker production but did stimulate production of reactive oxygen species (ROS), an effect that could be reversed by subsequent treatment with the Fpr2 ligand QC1. Further investigation revealed that oAß-induced ROS production was associated with NADPH oxidase activation and a shift in BV2 cell metabolic phenotype, activating the pentose phosphate pathway and NADPH production, changes that were again reversed by QC1 treatment. Microglial oAß-stimulated ROS production was sufficient to induce apoptosis of bystander SH-SY5Y cells, an effect that could be prevented by QC1 treatment. In this study, we provide proof-of-concept data that indicate exploitation of the proresolving receptor Fpr2 can reverse damaging oAß-induced microglial activation. Future strategies that are aimed at restraining neuroinflammation in conditions such as AD should examine proresolving actors as a mechanism to harness the brain's endogenous healing pathways and limit neuroinflammatory damage.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Microglía/patología , Receptores de Formil Péptido/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Antioxidantes/metabolismo , Línea Celular , Respiración de la Célula/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Inflamación/patología , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Vía de Pentosa Fosfato/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Formil Péptido/agonistas
5.
Eur J Pharmacol ; 872: 172971, 2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-32004526

RESUMEN

Human melanocortin MC1 and MC3 receptors expressed on C-20/A4 chondrocytes exhibit chondroprotective and anti-inflammatory effects when activated by melanocortin peptides. Nearly 9 million people in the UK suffer from osteoarthritis, and bacterial infections play a role in its development. Here, we evaluate the effect of a panel of melanocortin peptides with different selectivity for human melanocortin MC1 (α-MSH, BMS-470539 dihydrochloride) and MC3 ([DTrp8]-γ-MSH, PG-990) receptors and C-terminal peptide α-MSH11-13(KPV), on inhibiting LPS-induced chondrocyte death, pro-inflammatory mediators and induction of anti-inflammatory proteins. C-20/A4 chondrocytes were treated with a panel of melanocortin peptides prophylactically and therapeutically in presence of LPS (0.1 µg/ml). The chondroprotective properties of these peptides determined by cell viability assay, RT-PCR, ELISA for detection of changes in inflammatory markers (IL-6, IL-8 and MMP-1, -3 and -13) and western blotting for expression of the anti-inflammatory protein heme-oxygenase-1. C-20/A4 expressed human melanocortin MC1 and MC3 receptors and melanocortin peptides elevated cAMP. LPS stimulation caused a reduction in C-20/A4 viability, attenuated by the human melanocortin MC1 receptor agonist BMS-470539 dihydrochloride, and MC3 receptor agonists PG-990 and [DTrp8]-γ-MSH. Prophylactic and therapeutic regimes of [DTrp8]-γ-MSH significantly inhibited LPS-induced modulation of cartilage-damaging IL-6, IL-8, MMPs -1,-3 and -13 mediators both prophylactically and therapeutically, whilst human melanocortin MC1 and MC3 receptor agonists promoted an increase in HO-1 production. In the presence of LPS, activation of human melanocortin MC1 and MC3 receptors provided potent chondroprotection, upregulation of anti-inflammatory proteins and downregulation of inflammatory and proteolytic mediators involved in cartilage degradation, suggesting a new avenue for osteoarthritis treatment.


Asunto(s)
Antiinflamatorios/farmacología , Condrocitos/efectos de los fármacos , Receptor de Melanocortina Tipo 1/agonistas , Receptor de Melanocortina Tipo 3/agonistas , Línea Celular , Condrocitos/inmunología , Condrocitos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Humanos , Imidazoles , Lipopolisacáridos/inmunología , Osteoartritis/tratamiento farmacológico , Osteoartritis/inmunología , Osteoartritis/patología
6.
Pharmacol Ther ; 192: 170-187, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30081050

RESUMEN

Extracellular vesicles (EVs) are a heterogenous population of vesicles originate from cells. EVs are found in different biofluids and carry different macromolecules, including proteins, lipids, and nucleic acids, providing a snap shot of the parental cells at the time of release. EVs have the ability to transfer molecular cargoes to other cells and can initiate different physiological and pathological processes. Mounting lines of evidence demonstrated that EVs' cargo and machinery is affected in disease states, positioning EVs as potential sources for the discovery of novel biomarkers. In this review, we demonstrate a conceptual overview of the EV field with particular focus on their nucleic acid cargoes. Current knowledge of EV subtypes, nucleic acid cargo and pathophysiological roles are outlined, with emphasis placed on advantages against competing analytes. We review the utility of EVs and their nucleic acid cargoes as biomarkers and critically assess the newly available advances in the field of EV biomarkers and high throughput technologies. Challenges to achieving the diagnostic potential of EVs, including sample handling, EV isolation, methodological considerations, and bioassay reproducibility are discussed. Future implementation of 'omics-based technologies and integration of systems biology approaches for the development of EV-based biomarkers and personalized medicine are also considered.


Asunto(s)
Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Ácidos Nucleicos/metabolismo , Animales , Humanos , Medicina de Precisión , Proteínas/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 35(9): 1936-44, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26112010

RESUMEN

OBJECTIVE: Neutrophil recruitment is a key process in the pathogenesis of stroke, and may provide a valuable therapeutic target. Targeting the melanocortin (MC) receptors has previously shown to inhibit leukocyte recruitment in peripheral inflammation, however, it is not known whether treatments are effective in the unique cerebral microvascular environment. Here, we provide novel research highlighting the effects of the MC peptides on cerebral neutrophil recruitment, demonstrating important yet discrete roles for both MC1 and MC3. APPROACH AND RESULTS: Using intravital microscopy, in 2 distinct murine models of cerebral ischemia-reperfusion (I/R) injury, we have investigated MC control for neutrophil recruitment. After global I/R, pharmacological treatments suppressed pathological neutrophil recruitment. MC1 selective treatment rapidly inhibited neutrophil recruitment while a nonselective MC agonist provided protection even when coadministered with an MC3/4 antagonist, suggesting the importance of early MC1 signaling. However, by 2-hour reperfusion, MC1-mediated effects were reduced, and MC3 anti-inflammatory circuits predominated. Mice bearing a nonfunctional MC1 displayed a transient exacerbation of neutrophil recruitment after global I/R, which diminished by 2 hours. However importantly, enhanced inflammatory responses in both MC1 mutant and MC3 (-/-) mice resulted in increased infarct size and poor functional outcome after focal I/R. Furthermore, we used an in vitro model of leukocyte recruitment to demonstrate these anti-inflammatory actions are also effective in human cells. CONCLUSIONS: These studies reveal for the first time MC control for neutrophil recruitment in the unique pathophysiological context of cerebral I/R, while also demonstrating the potential therapeutic value of targeting multiple MCs in developing effective therapeutics.


Asunto(s)
Isquemia Encefálica/prevención & control , Regulación de la Expresión Génica , Infiltración Neutrófila/genética , ARN Mensajero/genética , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 3/genética , Daño por Reperfusión/complicaciones , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Receptor de Melanocortina Tipo 1/antagonistas & inhibidores , Receptor de Melanocortina Tipo 1/biosíntesis , Receptor de Melanocortina Tipo 3/antagonistas & inhibidores , Receptor de Melanocortina Tipo 3/biosíntesis , Daño por Reperfusión/metabolismo
8.
Biochem Pharmacol ; 92(2): 336-47, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25175737

RESUMEN

INTRODUCTION: Mechanical injury can greatly influence articular cartilage, propagating inflammation, cell injury and death - risk factors for the development of osteoarthritis. Melanocortin peptides and their receptors mediate anti-inflammatory and pro-resolving mechanisms in chondrocytes. This study aimed to investigate the potential chondroprotective properties of α-MSH and [DTRP(8)]-γ-MSH in mechanically injured cartilage explants, their ability to inhibit pro-inflammatory and stimulate anti-inflammatory cytokines in in situ and in freshly isolated articular chondrocytes. METHODS: The effect of melanocortins on in situ chondrocyte viability was investigated using confocal laser scanning microscopy of bovine articular cartilage explants, subjected to a single blunt impact (1.14N, 6.47 kPa) delivered by a drop tower. Chondroprotective effects of α-MSH, [DTRP(8)]-γ-MSH and dexamethasone on cytokine release by TNF-α-activated freshly isolated articular chondrocytes/mechanically injured cartilage explants were investigated by ELISA. RESULTS: A single impact to cartilage caused discreet areas of chondrocyte death, accompanied by pro-inflammatory cytokine release; both parameters were modulated by α-MSH, [DTRP(8)]-γ-MSH and dexamethasone. Melanocortin pre-treatment of TNF-α-stimulated freshly isolated chondrocytes resulted in a bell-shaped inhibition in IL-1ß, IL-6 and IL-8, and elevation of IL-10 production. The MC3/4 antagonist, SHU9119, abrogated the effect of [DTRP(8)]-γ-MSH but not α-MSH on cytokine release. CONCLUSION: Melanocortin peptide pre-treatment prevented chondrocyte death following mechanical impact to cartilage and led to a marked reduction of pro-inflammatory cytokines, whilst prompting the production of anti-inflammatory/pro-resolving cytokine IL-10. Development of small molecule agonists towards melanocortin receptors could thus be a viable approach for preventing chondrocyte inflammation and death within cartilage and represent an alternative approach for the treatment of osteoarthritis.


Asunto(s)
Cartílago Articular/lesiones , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Mediadores de Inflamación/metabolismo , Fenómenos Mecánicos , Melanocortinas/farmacología , Animales , Cartílago Articular/efectos de los fármacos , Bovinos , Células Cultivadas , Fenómenos Mecánicos/efectos de los fármacos , Técnicas de Cultivo de Órganos , alfa-MSH/farmacología
9.
FASEB J ; 26(12): 4977-89, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22964301

RESUMEN

Unregulated inflammation underlies many diseases, including sepsis. Much interest lies in targeting anti-inflammatory mechanisms to develop new treatments. One such target is the anti-inflammatory protein annexin A1 (AnxA1) and its receptor, FPR2/ALX. Using intravital videomicroscopy, we investigated the role of AnxA1 and FPR2/ALX in a murine model of endotoxin-induced cerebral inflammation [intraperitoneal injection of lipopolysaccharide (LPS)]. An inflammatory response was confirmed by elevations in proinflammatory serum cytokines, increased cerebrovascular permeability, elevation in brain myeloperoxidase, and increased leukocyte rolling and adhesion in cerebral venules of wild-type (WT) mice, which were further exacerbated in AnxA1-null mice. mRNA expression of TLR2, TLR4, MyD-88, and Ly96 was also assessed. The AnxA1-mimetic peptide, AnxA1(Ac2-26) (100 µg/mouse, ∼33 µmol) mitigated LPS-induced leukocyte adhesion in WT and AnxA1-null animals without affecting leukocyte rolling, in comparison to saline control. AnxA1(Ac2-26) effects were attenuated by Boc2 (pan-FPR antagonist, 10 µg/mouse, ∼12 nmol), and by minocycline (2.25 mg/mouse, ∼6.3 nmol). The nonselective Fpr agonists, fMLP (6 µg/mouse, ∼17 nmol) and AnxA1(Ac2-26), and the Fpr2-selective agonist ATLa (5 µg/mouse, ∼11 nmol) were without effect in Fpr2/3(-/-) mice. In summary, our novel results demonstrate that the AnxA1/FPR2 system has an important role in effecting the resolution of cerebral inflammation in sepsis and may, therefore, provide a novel therapeutic target.


Asunto(s)
Anexina A1/metabolismo , Encéfalo/metabolismo , Inflamación/metabolismo , Leucocitos/metabolismo , Receptores de Formil Péptido/metabolismo , Sepsis/metabolismo , Animales , Anexina A1/química , Anexina A1/genética , Encéfalo/irrigación sanguínea , Encéfalo/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Circulación Cerebrovascular/efectos de los fármacos , Citocinas/sangre , Expresión Génica/efectos de los fármacos , Inflamación/sangre , Inflamación/inducido químicamente , Inyecciones Intraperitoneales , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/genética , Leucocitos/patología , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/toxicidad , Antígeno 96 de los Linfocitos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía por Video , Minociclina/farmacología , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Toll-Like/genética
10.
Microsc Res Tech ; 75(7): 991-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22514026

RESUMEN

We present three novel reproducible methodologies for the quantification of changes in actin organization from microscope images. Striation and integrative analysis were devised for the investigation of trans-cellular filaments and F-actin localization, respectively, in response to physiological or mechanical actin-modulatory conditions. Additionally, the Parker-Qusous (PQ) formula was developed as a measure of total quantity of F-actin, independent of cell volume changes, whereby fluorescence intensity was divided by the cube root of cell volume, squared. Values obtained were quantified in Mauricean Units (Mu; pixel/µm(3)). Upon isolation, there was a 49% decrease in total F-actin fluorescence from 1.91 ± 0.16 pixel/µm(3) (Mu) to 0.95 ± 0.55 Mu, whereas upon culture, an apparent increase in total fluorescence was deemed insignificant due to an increase in average cell volume, with a rise, however, in striation units (StU) from 1 ± 1 to 5 ± 1 StU/cell, and a decrease in percentage cortical fluorescence to 30.45% ± 1.52% (P = 7.8 × 10(-5)). Freshly isolated chondrocytes exhibited a decrease in total F-actin fluorescence to 0.61 ± 0.05 Mu and 0.32 ± 0.02 Mu, 10 min posthypertonic and hypotonic challenges, respectively. Regulatory volume decrease was inhibited in the presence of REV5901 with maintenance of actin levels at 1.15 Mu. Following mechanical impact in situ, there was a reduction in total F-actin fluorescence to 0.95 ± 0.08 Mu and 0.74 ± 0.06 Mu under isotonic and hypotonic conditions, respectively, but not under hypertonic conditions. We report simple methodologies for quantification of changes in actin organization, which will further our understanding of the role of actin in various cellular stress responses. These techniques can be applied to better quantify changes in localization of various proteins using fluorescent labeling.


Asunto(s)
Actinas/análisis , Condrocitos/química , Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Fluorescente/métodos , Animales , Bovinos , Modelos Teóricos
11.
Br J Pharmacol ; 167(1): 67-79, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22471953

RESUMEN

BACKGROUND AND PURPOSE: Melanocortin MC(1) and MC(3 ) receptors, mediate the anti-inflammatory effects of melanocortin peptides. Targeting these receptors could therefore lead to development of novel anti-inflammatory therapeutic agents. We investigated the expression of MC(1) and MC(3) receptors on chondrocytes and the role of α-melanocyte-stimulating hormone (α-MSH) and the selective MC(3) receptor agonist, [DTRP(8) ]-γ-MSH, in modulating production of inflammatory cytokines, tissue-destructive proteins and induction of apoptotic pathway(s) in the human chondrocytic C-20/A4 cells. EXPERIMENTAL APPROACH: Effects of α-MSH, [DTRP(8) ]-γ-MSH alone or in the presence of the MC(3/4) receptor antagonist, SHU9119, on TNF-α induced release of pro-inflammatory cytokines, MMPs, apoptotic pathway(s) and cell death in C-20/A4 chondrocytes were investigated, along with their effect on the release of the anti-inflammatory cytokine IL-10. KEY RESULTS: C-20/A4 chondrocytes expressed functionally active MC(1,3) receptors. α-MSH and [DTRP(8) ]-γ-MSH treatment, for 30 min before TNF-α stimulation, provided a time-and-bell-shaped concentration-dependent decrease in pro-inflammatory cytokines (IL-1ß, IL-6 and IL-8) release and increased release of the chondroprotective and anti-inflammatory cytokine, IL-10, whilst decreasing expression of MMP1, MMP3, MMP13 genes.α-MSH and [DTRP(8) ]-γ-MSH treatment also inhibited TNF-α-induced caspase-3/7 activation and chondrocyte death. The effects of [DTRP(8) ]-γ-MSH, but not α-MSH, were abolished by the MC(3/4) receptor antagonist, SHU9119. CONCLUSION AND IMPLICATIONS: Activation of MC(1) /MC(3) receptors in C-20/A4 chondrocytes down-regulated production of pro-inflammatory cytokines and cartilage-destroying proteinases, inhibited initiation of apoptotic pathways and promoted release of chondroprotective and anti-inflammatory cytokines. Developing small molecule agonists to MC(1) /MC(3) receptors could be a viable approach for developing chondroprotective and anti-inflammatory therapies in rheumatoid and osteoarthritis.


Asunto(s)
Antiinflamatorios/farmacología , Condrocitos/efectos de los fármacos , Hormonas Estimuladoras de los Melanocitos/farmacología , Sustancias Protectoras/farmacología , alfa-MSH/farmacología , gamma-MSH/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Condrocitos/metabolismo , AMP Cíclico/metabolismo , Citocinas/metabolismo , Dinoprostona/metabolismo , Humanos , Metaloproteinasas de la Matriz/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 3/metabolismo
12.
Cells Tissues Organs ; 196(3): 251-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22398355

RESUMEN

In cartilage research bovine articular cartilage is used as an alternative to human tissue. However, animal material is subject to availability and primary cultures undergo senescence, limiting their use. Here we report the immortalization of primary bovine chondrocytes, which could be used as a surrogate for freshly isolated chondrocytes. Chondrocytes were isolated from cartilage explants and immortalized using 1.0 µg/ml benzo[alpha]pyrene. For 3-dimensional culture, chondrocytes were resuspended in 0.5% low-melt agarose at high density (HD) and cultured for 24 h prior to determining changes in expression profile and morphology. A13/BACii chondrocytes acquired a 'flat' irregular morphology and a foetal-like cell volume (1,509.59 ± 182.04 µm(3)). The human cell line C-20/A4 showed a statistically similar volume and length to A13/BACii. Two-dimensional-cultured A13/BACii expressed elevated levels of type I collagen (col1), reduced levels of type II collagen (col2) compared to freshly isolated chondrocytes and an overall col2 to col1 expression ratio (col2:col1) of 0.11 ± 0.01. Upon 3-dimensional encapsulation, there was a significant rise in col2 expression in both A13/BACii and C-20/A4, suggesting a capacity for redifferentiation in both cell lines with a return of col2:col1 values of A13/BACii to values previously observed in primary chondrocytes. A13/BACii chondrocytes expressed aggrecan, matrix metalloproteinase (MMP)-3, MMP-9 and MMP-13, further supporting indications of the differentiated phenotype. Here we report the creation of a novel chondrocytic cell line and demonstrate its strong potential for redifferentiation upon HD 3-dimensional encapsulation, providing an alternative to conventional dedifferentiated cell lines and primary culture.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Agrecanos/metabolismo , Animales , Benzo(a)pireno/farmacología , Biomarcadores/metabolismo , Bioprótesis , Cartílago Articular/efectos de los fármacos , Cartílago Articular/fisiología , Bovinos , Técnicas de Cultivo de Célula , Desdiferenciación Celular/efectos de los fármacos , Desdiferenciación Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Transformada , Condrocitos/efectos de los fármacos , Condrocitos/fisiología , Colágeno Tipo II/metabolismo , Metaloproteasas/metabolismo , Microscopía Confocal , Fenotipo , Costillas/citología , Sefarosa , Rodilla de Cuadrúpedos
13.
Trends Pharmacol Sci ; 32(2): 90-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21185610

RESUMEN

The melanocortin receptors are a subfamily of G-protein-coupled, rhodopsin-like receptors that are rapidly being acknowledged as an extremely promising target for pharmacological intervention in a variety of different inflammatory pathologies, including stroke. Stroke continues to be a leading cause of death worldwide, with risk factors including smoking, diabetes, hypertension and obesity. The pathophysiology of stroke is highly complex: reintroduction of blood flow to the infarcted brain region is paramount in limiting ischaemic damage caused by stroke, yet a concomitant inflammatory response can compound tissue damage. The possibilities of pro-resolving treatments that target this inflammatory response have only recently begun to be explored. This review discusses the endogenous roles of the melanocortin system in reducing characterized aspects of inflammation, and how these, together with potent neuroprotective actions, suggest its potential as a therapeutic target in stroke.


Asunto(s)
Terapia Molecular Dirigida , Receptores de Melanocortina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Humanos , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
14.
FASEB J ; 24(12): 4835-43, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20702773

RESUMEN

The development of biological therapies has improved management of rheumatoid arthritis. However, costs and unresponsiveness to therapy in a sizeable proportion of patients limit their use, making it imperative to identify new targets for drug development programs. Here we investigated the melanocortin-receptor type 3 (MC(3)) pathway. Gene-deficient mice were subjected to a model of serum-transfer-induced arthritis and joints analyzed for gene expression (cytokines, MCs) and morphology. Pharmacological analyses were also conducted in this model. Osteoclastogenesis was studied from bone marrow cells. Mc(3)(-/-) mice displayed an exacerbated inflammatory arthritis, associated with prominent bone erosion and higher articular expression of Rankl. Osteoclastogenesis studied from Mc(3)(-/-) bone marrow cells revealed a higher degree of responsiveness to Rankl, linked to prolonged NF-κB activation compared to wild types. Up-regulation of a discrete set of inflammatory genes, including Il-1ß, Il-6, and Nos2, was measured in Mc(3)(-/-) mice, and a marked up-regulation of joint Mc(3) accompanied arthritis resolution in wild-type mice. Administration of an MC(3) agonist, D[Trp8]-γ-MSH, attenuated disease incidence and severity in wild-type but not Mc(3)(-/-) mice. Overall, these findings identify MC(3)-mediated signaling as a beneficial pathway in experimental arthritis; hence this receptor is a novel target for the development of therapeutics for arthritis.


Asunto(s)
Artritis Experimental/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Receptor de Melanocortina Tipo 3/metabolismo , Animales , Artritis Experimental/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteogénesis/genética , Osteogénesis/fisiología , Reacción en Cadena de la Polimerasa , Receptor de Melanocortina Tipo 3/genética
15.
Arterioscler Thromb Vasc Biol ; 30(9): 1718-24, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20558817

RESUMEN

OBJECTIVE: To determine whether the inhibitory action of the antiallergic cromone "mast cell stabilizing" drugs on polymorphonuclear leukocyte (PMN) trafficking is mediated through an annexin-A1 (Anx-A1) dependent mechanism. METHODS AND RESULTS: Intravital microscopy was used to monitor the actions of cromones in the inflamed microcirculation. Reperfusion injury provoked a dramatic increase in adherent and emigrated leukocytes in the mesenteric vascular bed, associated with augmented tissue levels of myeloperoxidase. Nedocromil, 2 to 20 mg/kg, significantly (P<0.05) inhibited cell adhesion and emigration, as well as myeloperoxidase release, in wild-type but not Anx-A1(-/-) mice. Short pretreatment of human PMNs with nedocromil, 10 nmol/L, inhibited cell adhesion (P<0.05) in the flow chamber assay, and this effect was reversed by specific anti-AnxA1 or a combination of antiformyl peptide receptors 1 and 2, but not irrelevant control, antibodies. Western blotting experiments revealed that cromones stimulate protein kinase C-dependent phosphorylation and release Anx-A1 in human PMNs. CONCLUSIONS: We propose a novel mechanism to explain the antiinflammatory actions of cromones on PMN trafficking, an effect that has long puzzled investigators.


Asunto(s)
Anexina A1/metabolismo , Antialérgicos/farmacología , Antiinflamatorios/farmacología , Adhesión Celular/efectos de los fármacos , Cromolin Sódico/farmacología , Células Endoteliales/efectos de los fármacos , Nedocromil/farmacología , Neutrófilos/efectos de los fármacos , Animales , Anexina A1/deficiencia , Anexina A1/genética , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Rodamiento de Leucocito/efectos de los fármacos , Masculino , Oclusión Vascular Mesentérica/tratamiento farmacológico , Oclusión Vascular Mesentérica/inmunología , Oclusión Vascular Mesentérica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/efectos de los fármacos , Microscopía por Video , Neutrófilos/inmunología , Neutrófilos/metabolismo , Peritonitis/tratamiento farmacológico , Peritonitis/inmunología , Peritonitis/metabolismo , Peroxidasa/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Transporte de Proteínas , Receptores de Formil Péptido/efectos de los fármacos , Receptores de Formil Péptido/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Factores de Tiempo
16.
ScientificWorldJournal ; 10: 434-56, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20305986

RESUMEN

Saliva has been described as the mirror of the body. In a world of soaring healthcare costs and an environment where rapid diagnosis may be critical to a positive patient outcome, saliva is emerging as a viable alternative to blood sampling. In this review, we discuss the composition and various physiological roles of saliva in the oral cavity, including soft tissue protection, antimicrobial activities, and oral tissue repair. We then explore saliva as a diagnostic marker of local oral disease and focus particularly on oral cancers. The cancer theme continues when we focus on systemic disease diagnosis from salivary biomarkers. Communicable disease is the focus of the next section where we review the literature relating to the direct and indirect detection of pathogenic infections from human saliva. Finally, we discuss hormones involved in appetite regulation and whether saliva is a viable alternative to blood in order to monitor hormones that are involved in satiety.


Asunto(s)
Diagnóstico , Saliva/fisiología , Biomarcadores/análisis , Humanos , Saliva/inmunología
17.
Pulm Pharmacol Ther ; 21(6): 866-73, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18992358

RESUMEN

In this study we set out to ascertain whether melanocortin peptides could be potential therapeutic agents in allergic and non-allergic models of lung inflammation by identifying the receptor(s) involved using a molecular, genetic and pharmacological approach. Western blot analyses revealed expression of the melanocortin receptor (MCR) type 1 and 3 on alveolar macrophages from wild-type mice. Alveolar macrophage incubation, with the selective MC3R agonist [D-TRP(8)]-gamma-MSH and pan-agonist alpha-MSH but not the selective MC1R agonist MS05, led to an increase in cAMP in wild-type macrophages. This increase occurred also in macrophages taken from recessive yellow (e/e; bearing a mutant and inactive MC1R) mice but not from MC3R-null mice. In an allergic model of inflammation, the pan-agonist alpha-MSH and selective MC3R agonist [D-TRP(8)]-gamma-MSH displayed significant attenuation of both eosinophil and lymphocyte accumulation but not IL-5 levels in wild-type and recessive yellow e/e mice. However in MC3R-null mice, alpha-MSH failed to cause a significant inhibition in these parameters, highlighting a preferential role for MC3R in mediating the anti-inflammatory effects of melanocortins in this model. Utilising a non-allergic model of LPS-induced lung neutrophilia, the pan-agonist alpha-MSH and selective MC3R agonist [D-TRP(8)]-gamma-MSH displayed significant attenuation of neutrophil accumulation and inhibition of TNF-alpha release. Thus, this study highlights that melanocortin peptides inhibit leukocyte accumulation in a model of allergic and non-allergic inflammation and this protective effect is associated with activation of the MC3R. The inhibition of leukocyte accumulation is via inhibition of TNF-alpha in the non-allergic model of inflammation but not IL-5 in the allergic model. These data have highlighted the potential for selective MC3R agonists as novel anti-inflammatory therapeutics in lung inflammation.


Asunto(s)
Neumonía/metabolismo , Receptor de Melanocortina Tipo 3/fisiología , alfa-MSH/farmacología , gamma-MSH/farmacología , Animales , Antiinflamatorios/farmacología , Eosinófilos/metabolismo , Interleucina-5/química , Linfocitos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/fisiología , Melanocortinas/farmacología , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Péptidos/farmacología , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Receptor de Melanocortina Tipo 1/fisiología , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 3/genética , Receptor de Melanocortina Tipo 3/metabolismo , Factor de Necrosis Tumoral alfa/química , alfa-MSH/agonistas , gamma-MSH/agonistas
18.
FASEB J ; 22(12): 4228-38, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18757499

RESUMEN

The existence of anti-inflammatory circuits centered on melanocortin receptors (MCRs) has been supported by the inhibitory properties displayed by melanocortin peptides in models of inflammation and tissue injury. Here we addressed the pathophysiological effect that one MCR, MCR type 3 (MC3R), might have on vascular inflammation. After occlusion (35 min) and reopening of the superior mesenteric artery, MC3R-null mice displayed a higher degree of plasma extravasation (45 min postreperfusion) and cell adhesion and emigration (90 min postreperfusion). These cellular alterations were complemented by higher expression of mesenteric tissue CCL2 and CXCL1 (mRNA and protein) and myeloperoxydase, as compared with wild-type animals. MC1R and MC3R mRNA and protein were both expressed in the inflamed mesenteric tissue; however, no changes in vascular responses were observed in a mouse colony bearing an inactive MC1R. Pharmacological treatment of animals with a selective MC3R agonist ([D-Trp(8)]-gamma-melanocyte-stimulating hormone; 10 microg i.v.) produced marked attenuation of cell adhesion, emigration, and chemokine generation; such effects were absent in MC3R-null mice. These new data reveal the existence of a tonic inhibitory signal provided by MC3R in the mesenteric microcirculation of the mouse, acting to down-regulate cell trafficking and local mediator generation.


Asunto(s)
Inflamación/fisiopatología , Mesenterio/irrigación sanguínea , Microcirculación/fisiología , Receptor de Melanocortina Tipo 3/deficiencia , Daño por Reperfusión/fisiopatología , Animales , Antígeno CD11b/biosíntesis , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/biosíntesis , Quimiocina CXCL1/biosíntesis , Regulación hacia Abajo , Mutación del Sistema de Lectura , Mediadores de Inflamación/fisiología , Selectina L/biosíntesis , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Mesenterio/metabolismo , Ratones , Fenotipo , Receptor de Melanocortina Tipo 3/agonistas , Receptor de Melanocortina Tipo 3/genética
19.
ScientificWorldJournal ; 7: 1463-1464, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-29861697
20.
FASEB J ; 20(13): 2234-41, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17077300

RESUMEN

In this study we have characterized the anti-inflammatory profile of a selective melanocortin type 3 receptor (MC3-R) ligand [D-Trp8]-gamma-MSH, validating in vitro results with analyses in mice deficient for this receptor subtype. In wild-type (WT) macrophages, [D-Trp8]-gamma-MSH activated MC3-R (as tested by accumulation of cyclic AMP) and inhibited (approximately 50%) the release of interleukin (IL)-1 and the chemokine KC (CXCL1), but was ineffective in cells taken from MC3-R null mice. In vivo, administration of 3-30 microg [D-Trp8]-gamma-MSH significantly inhibited leukocyte influx and cytokine production in a model of crystal-induced peritonitis, and these effects were absent in MC3-R null mice or blocked by coadministration of an MC3-R antagonist. Finally, in a model of gouty arthritis, direct injection of urate crystals into the rat joint provoked a marked inflammatory reaction that was significantly inhibited (approximately 70%) by systemic or local administration of [D-Trp8]-gamma-MSH. In conclusion, using an integrated transgenic and pharmacological approach, we provide strong proof of concept for the development of selective MC3-R agonists as novel anti-inflammatory therapeutics.


Asunto(s)
Artritis Gotosa/fisiopatología , Inflamación/prevención & control , Macrófagos Peritoneales/fisiología , Receptor de Melanocortina Tipo 3/fisiología , Receptores de Melanocortina/fisiología , Animales , Artritis Experimental/fisiopatología , Artritis Experimental/prevención & control , Artritis Gotosa/inducido químicamente , Artritis Gotosa/prevención & control , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 3/antagonistas & inhibidores , Receptor de Melanocortina Tipo 3/deficiencia , Receptor de Melanocortina Tipo 3/genética , Receptores de Melanocortina/genética , Ácido Úrico/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...