Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int J Mol Sci ; 24(21)2023 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-37958978

RESUMO

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.


Assuntos
Lipopolissacarídeos , Microglia , Animais , Camundongos , Anti-Inflamatórios/farmacologia , Morte Celular , Lipopolissacarídeos/farmacologia , Microglia/metabolismo , NF-kappa B/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 35(9): 1936-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26112010

RESUMO

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.


Assuntos
Isquemia Encefálica/prevenção & controle , Regulação da Expressão Gênica , Infiltração de Neutrófilos/genética , RNA Mensageiro/genética , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 3 de Melanocortina/genética , Traumatismo por Reperfusão/complicações , Animais , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos , Receptor Tipo 1 de Melanocortina/antagonistas & inibidores , Receptor Tipo 1 de Melanocortina/biossíntese , Receptor Tipo 3 de Melanocortina/antagonistas & inibidores , Receptor Tipo 3 de Melanocortina/biossíntese , Traumatismo por Reperfusão/metabolismo
3.
FASEB J ; 26(12): 4977-89, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964301

RESUMO

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.


Assuntos
Anexina A1/metabolismo , Encéfalo/metabolismo , Inflamação/metabolismo , Leucócitos/metabolismo , Receptores de Formil Peptídeo/metabolismo , Sepse/metabolismo , Animais , Anexina A1/química , Anexina A1/genética , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Circulação Cerebrovascular/efeitos dos fármacos , Citocinas/sangue , Expressão Gênica/efeitos dos fármacos , Inflamação/sangue , Inflamação/induzido quimicamente , Injeções Intraperitoneais , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Migração e Rolagem de Leucócitos/genética , Leucócitos/patologia , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/toxicidade , Antígeno 96 de Linfócito/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Vídeo , Minociclina/farmacologia , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptores de Formil Peptídeo/antagonistas & inibidores , Receptores de Formil Peptídeo/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/genética
4.
Cells Tissues Organs ; 196(3): 251-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22398355

RESUMO

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.


Assuntos
Cartilagem Articular/citologia , Condrócitos/citologia , Agrecanas/metabolismo , Animais , Benzo(a)pireno/farmacologia , Biomarcadores/metabolismo , Bioprótese , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/fisiologia , Bovinos , Técnicas de Cultura de Células , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Colágeno Tipo II/metabolismo , Metaloproteases/metabolismo , Microscopia Confocal , Fenótipo , Costelas/citologia , Sefarose , Joelho de Quadrúpedes
5.
FEBS J ; 289(7): 1801-1822, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33811735

RESUMO

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.


Assuntos
Doença de Alzheimer , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/metabolismo , Idoso , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Humanos , Microglia/metabolismo , Receptores de Formil Peptídeo/genética
6.
FASEB J ; 24(12): 4835-43, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20702773

RESUMO

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.


Assuntos
Artrite Experimental/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Receptor Tipo 3 de Melanocortina/metabolismo , Animais , Artrite Experimental/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese/genética , Osteogênese/fisiologia , Reação em Cadeia da Polimerase , Receptor Tipo 3 de Melanocortina/genética
7.
Arterioscler Thromb Vasc Biol ; 30(9): 1718-24, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558817

RESUMO

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.


Assuntos
Anexina A1/metabolismo , Antialérgicos/farmacologia , Anti-Inflamatórios/farmacologia , Adesão Celular/efeitos dos fármacos , Cromolina Sódica/farmacologia , Células Endoteliais/efeitos dos fármacos , Nedocromil/farmacologia , Neutrófilos/efeitos dos fármacos , Animais , Anexina A1/deficiência , Anexina A1/genética , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Migração e Rolagem de Leucócitos/efeitos dos fármacos , Masculino , Oclusão Vascular Mesentérica/tratamento farmacológico , Oclusão Vascular Mesentérica/imunologia , Oclusão Vascular Mesentérica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/efeitos dos fármacos , Microscopia de Vídeo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peritonite/tratamento farmacológico , Peritonite/imunologia , Peritonite/metabolismo , Peroxidase/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Transporte Proteico , Receptores de Formil Peptídeo/efeitos dos fármacos , Receptores de Formil Peptídeo/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/metabolismo , Fatores de Tempo
8.
Nat Med ; 8(11): 1296-302, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12368905

RESUMO

Aspirin (ASA) and dexamethasone (DEX) are widely used anti-inflammatory agents yet their mechanism(s) for blocking polymorphonuclear neutrophil (PMN) accumulation at sites of inflammation remains unclear. Here, we report that inhibition of PMN infiltration by ASA and DEX is a property shared by aspirin-triggered lipoxins (ATL) and the glucocorticoid-induced annexin 1 (ANXA1)-derived peptides that are both generated in vivo and act at the lipoxin A(4) receptor (ALXR/FPRL1) to halt PMN diapedesis. These structurally diverse ligands specifically interact directly with recombinant human ALXR demonstrated by specific radioligand binding and function as well as immunoprecipitation of PMN receptors. In addition, the combination of both ATL and ANXA1-derived peptides limited PMN infiltration and reduced production of inflammatory mediators (that is, prostaglandins and chemokines) in vivo. Together, these results indicate functional redundancies in endogenous lipid and peptide anti-inflammatory circuits that are spatially and temporally separate, where both ATL and specific ANXA1-derived peptides act in concert at ALXR to downregulate PMN recruitment to inflammatory loci.


Assuntos
Anti-Inflamatórios/farmacologia , Aspirina/farmacologia , Dexametasona/farmacologia , Metabolismo dos Lipídeos , Peptídeos/metabolismo , Receptores de Superfície Celular/agonistas , Receptores de Formil Peptídeo , Receptores de Lipoxinas , Animais , Linhagem Celular , Células Cultivadas , Humanos , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Ativação de Neutrófilo
9.
Cells ; 11(1)2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-35011694

RESUMO

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.


Assuntos
Diferenciação Celular , Osteoclastos/citologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Artrite Reumatoide/complicações , Diferenciação Celular/efeitos dos fármacos , Humanos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoporose/complicações , Transdução de Sinais/efeitos dos fármacos
10.
ScientificWorldJournal ; 10: 434-56, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20305986

RESUMO

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.


Assuntos
Diagnóstico , Saliva/fisiologia , Biomarcadores/análise , Humanos , Saliva/imunologia
11.
Oxid Med Cell Longev ; 2020: 2139192, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32617132

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Microglia/patologia , Receptores de Formil Peptídeo/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Antioxidantes/metabolismo , Linhagem Celular , Respiração Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Inflamação/patologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Via de Pentose Fosfato/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Formil Peptídeo/agonistas
12.
Eur J Pharmacol ; 872: 172971, 2020 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-32004526

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Condrócitos/efeitos dos fármacos , Receptor Tipo 1 de Melanocortina/agonistas , Receptor Tipo 3 de Melanocortina/agonistas , Linhagem Celular , Condrócitos/imunologia , Condrócitos/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Imidazóis , Lipopolissacarídeos/imunologia , Osteoartrite/tratamento farmacológico , Osteoartrite/imunologia , Osteoartrite/patologia
13.
FASEB J ; 22(12): 4228-38, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18757499

RESUMO

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.


Assuntos
Inflamação/fisiopatologia , Mesentério/irrigação sanguínea , Microcirculação/fisiologia , Receptor Tipo 3 de Melanocortina/deficiência , Traumatismo por Reperfusão/fisiopatologia , Animais , Antígeno CD11b/biossíntese , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/biossíntese , Quimiocina CXCL1/biossíntese , Regulação para Baixo , Mutação da Fase de Leitura , Mediadores da Inflamação/fisiologia , Selectina L/biossíntese , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Mesentério/metabolismo , Camundongos , Fenótipo , Receptor Tipo 3 de Melanocortina/agonistas , Receptor Tipo 3 de Melanocortina/genética
14.
Pharmacol Ther ; 192: 170-187, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30081050

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Ácidos Nucleicos/metabolismo , Animais , Humanos , Medicina de Precisão , Proteínas/metabolismo
15.
Pharmacol Ther ; 111(1): 1-15, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16488018

RESUMO

Adrenocorticotrophic hormone (ACTH(1-39)) and the melanocortins (alpha, beta and gamma-melanocyte-stimulating hormone [MSH]) are derived from a larger precursor molecule known as the pro-opiomelanocortin (POMC) protein. They exert their numerous biological effects by activating 7 transmembrane G-protein coupled receptors (GPCR), leading to adenylyl cyclase activation and subsequent cAMP accumulation within the target cell. To date, 5 melanocortin receptors (MCR) have been identified and termed MC1R to MC5R, they have been shown to have a wide and varied distribution throughout the body, being found in the central nervous system (CNS), periphery and immune cells. Melanocortins have a multitude of actions including: (i) modulating disease pathologies including arthritis, asthma, obesity; (ii) affecting functions, for example erectile dysfunction, skin tanning; and (iii) organ systems, for example cardiovascular system. Recently a mechanistic approach has been identified with alpha-MSH preventing NF-kappaB activation via the preservation and expression of IkappaBalphaprotein. This leads to a reduction of pro-inflammatory mediators including cytokines and inhibition of adhesion molecule expression, with subsequent reduction in leukocyte emigration. Development of selective ligands with an appropriate pharmacokinetic profile will enable a pharmacological evaluation of the potential beneficial effects of the melanocortins. In this review I have discussed the potential mechanistic action for the melanocortins and some of the disease pathologies shown to be modulated. This review proposes targeting the MCR with the ultimate aim of controlling many of the diseases that we face today.


Assuntos
Receptores de Melanocortina/metabolismo , Animais , Artrite Gotosa/tratamento farmacológico , Asma/tratamento farmacológico , Doenças Cardiovasculares/tratamento farmacológico , Desenho de Fármacos , Febre/tratamento farmacológico , Humanos , Inflamação/tratamento farmacológico , Doenças Inflamatórias Intestinais/tratamento farmacológico , Obesidade/tratamento farmacológico , Pró-Opiomelanocortina/metabolismo , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inibidores , Transdução de Sinais
16.
FASEB J ; 20(13): 2234-41, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17077300

RESUMO

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.


Assuntos
Artrite Gotosa/fisiopatologia , Inflamação/prevenção & controle , Macrófagos Peritoneais/fisiologia , Receptor Tipo 3 de Melanocortina/fisiologia , Receptores de Melanocortina/fisiologia , Animais , Artrite Experimental/fisiopatologia , Artrite Experimental/prevenção & controle , Artrite Gotosa/induzido quimicamente , Artrite Gotosa/prevenção & controle , Modelos Animais de Doenças , Regulação da Expressão Gênica , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptor Tipo 3 de Melanocortina/antagonistas & inibidores , Receptor Tipo 3 de Melanocortina/deficiência , Receptor Tipo 3 de Melanocortina/genética , Receptores de Melanocortina/genética , Ácido Úrico/toxicidade
17.
Peptides ; 27(2): 404-12, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16293345

RESUMO

Melanocortin peptides modulate cytokine release and adhesion molecule expression. Here we have investigated the early cell-signaling pathway responsible for the induction of interleukin-10 (IL-10) in RAW264.7 cells. Cell incubation with ACTH(1-39) or MTII (melanotan II) did not alter ERK1/2 and JNK phosphorylation, while p38 phosphorylation and intracellular cAMP accumulation occurred within minutes. ACTH(1-39) and MTII provoked a time-dependent accumulation of IL-10 that was abrogated by the PKA inhibitor H-89 and only partially blocked by the p38 MAPK inhibitor SB203580. Thus, in RAW264.7 cells, IL-10 induction by the melanocortins is via the PKA pathway, and this mechanism could contribute to their anti-inflammatory profile.


Assuntos
Interleucina-10/metabolismo , Macrófagos/fisiologia , Receptores de Melanocortina/fisiologia , Transdução de Sinais/fisiologia , Hormônio Adrenocorticotrópico/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Peptídeos Cíclicos/farmacologia , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
18.
Eur J Pharmacol ; 532(1-2): 138-44, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16442098

RESUMO

The clinically efficacious melanocortin peptide HP228 has here been investigated for its anti-inflammatory efficacy. In this study we have investigated the efficacy of HP228 in murine acute models of inflammation and myocardial ischaemia. Systemic treatment of mice with HP228 inhibited neutrophil accumulation in zymosan; urate crystal and carrageenan induced inflammatory models. In the urate model this was due to inhibition of pro-inflammatory chemokines and cytokines, whilst different mechanisms exist for zymosan peritonitis and carrageenan-induced air-pouch inflammation. HP228 was next evaluated in a model of myocardial ischaemia, another condition where cytokines and neutrophils are thought to play a causal role. HP228 caused a 50% reduction in myocardial damage following reperfusion. HP228 therefore inhibits the most important facet of the host inflammatory response namely leukocyte migration. These data show for the first time that the clinically efficacious peptide HP228 displays protective effects in models of inflammation and organ damage.


Assuntos
Inflamação/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oligopeptídeos/farmacologia , Doença Aguda , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Carragenina , Quimiocina CXCL1 , Quimiocinas , Quimiocinas CXC , AMP Cíclico/biossíntese , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Frequência Cardíaca/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-1/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Oligopeptídeos/uso terapêutico , Peritonite/induzido quimicamente , Peritonite/metabolismo , Peritonite/prevenção & controle , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Ácido Úrico , Zimosan , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
19.
ScientificWorldJournal ; 6: 1750-69, 2006 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17195873

RESUMO

Synthetic glucocorticoids are the most potent anti-inflammatory agents used to treat chronic inflammatory disease, such as asthma. However, a small number (<5%) of asthmatic patients and almost all patients with chronic obstructive pulmonary disease (COPD) do not respond well, or at all, to glucocorticoid therapy. If the molecular mechanism of glucocorticoid insensitivity is uncovered, it may in turn provide insight into the key mechanism of glucocorticoid action and allow a rational way to implement treatment regimens that restore glucocorticoid sensitivity. Glucocorticoids exert their effects by binding to a cytoplasmic glucocorticoid receptor (GR), which is subjected to post-translational modifications. Receptor phosphorylation, acetylation, nitrosylation, ubiquitinylation, and other modifications influence hormone binding, nuclear translocation, and protein half-life. Analysis of GR interactions to other molecules, such as coactivators or corepressors, may explain the genetic specificity of GR action. Priming with inflammatory cytokine or oxidative/nitrative stress is a mechanism for the glucocorticoid resistance observed in chronic inflammatory airway disease via reduction of corepressors or GR modification. Therapies targeting these aspects of the GR activation pathway may reverse glucocorticoid resistance in patients with glucocorticoid-insensitive airway disease and some patients with other inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease.


Assuntos
Anti-Inflamatórios/uso terapêutico , Asma/tratamento farmacológico , Asma/metabolismo , Glucocorticoides/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Receptores de Glucocorticoides/metabolismo , Asma/genética , Expressão Gênica , Humanos , Doença Pulmonar Obstrutiva Crônica/genética , Receptores de Glucocorticoides/genética
20.
Trends Pharmacol Sci ; 23(10): 447-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12368061

RESUMO

alpha-Melanocyte stimulating hormone (alpha-MSH) and other melanocortin peptides are potent anti-inflammatory agents that exhibit efficacy in many animal models of acute and chronic inflammation. These peptides are produced both centrally and peripherally, and exert their biological actions via activation of membrane-bound receptors. To exploit the therapeutic potential of such anti-inflammatory peptides, it is essential that the receptor subtype mediating these effects is identified with certainty.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Peptídeos/farmacologia , Receptores da Corticotropina/metabolismo , alfa-MSH/química , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Receptores da Corticotropina/efeitos dos fármacos , Receptores de Melanocortina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA