Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 5(2): 103030, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38678566

RESUMO

Gout is caused by the deposition of monosodium urate crystals (MSUc) in the joints, triggering a unique inflammatory and metabolic response in macrophages. Here, we present a protocol to generate MSUc for in vitro and in vivo studies in mouse and human cells. We describe steps for dissolving uric acid followed by crystallizing, purifying, evaluating, and analyzing MSUc. We then detail procedures for stimulating human/mouse-derived macrophages and determining endotoxin levels in MSUc preparation.


Assuntos
Cristalização , Gota , Macrófagos , Ácido Úrico , Ácido Úrico/metabolismo , Ácido Úrico/química , Animais , Humanos , Camundongos , Macrófagos/metabolismo , Gota/metabolismo
2.
Inflamm Res ; 73(5): 739-751, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38493256

RESUMO

OBJECTIVES: Cellular NAD+ declines in inflammatory states associated with increased activity of the leukocyte-expressed NADase CD38. In this study, we tested the potential role of therapeutically targeting CD38 and NAD+ in gout. METHODS: We studied cultured mouse wild type and CD38 knockout (KO) murine bone marrow derived macrophages (BMDMs) stimulated by monosodium urate (MSU) crystals and used the air pouch gouty inflammation model. RESULTS: MSU crystals induced CD38 in BMDMs in vitro, associated with NAD+ depletion, and IL-1ß and CXCL1 release, effects reversed by pharmacologic CD38 inhibitors (apigenin, 78c). Mouse air pouch inflammatory responses to MSU crystals were blunted by CD38 KO and apigenin. Pharmacologic CD38 inhibition suppressed MSU crystal-induced NLRP3 inflammasome activation and increased anti-inflammatory SIRT3-SOD2 activity in macrophages. BMDM RNA-seq analysis of differentially expressed genes (DEGs) revealed CD38 to control multiple MSU crystal-modulated inflammation pathways. Top DEGs included the circadian rhythm modulator GRP176, and the metalloreductase STEAP4 that mediates iron homeostasis, and promotes oxidative stress and NF-κB activation when it is overexpressed. CONCLUSIONS: CD38 and NAD+ depletion are druggable targets controlling the MSU crystal- induced inflammation program. Targeting CD38 and NAD+ are potentially novel selective molecular approaches to limit gouty arthritis.


Assuntos
ADP-Ribosil Ciclase 1 , Inflamação , Macrófagos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NAD , Ácido Úrico , Animais , ADP-Ribosil Ciclase 1/genética , ADP-Ribosil Ciclase 1/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Inflamação/tratamento farmacológico , Camundongos , NAD/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Células Cultivadas , Artrite Gotosa/tratamento farmacológico , Artrite Gotosa/metabolismo , Artrite Gotosa/genética , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos
3.
Arthritis Rheumatol ; 75(3): 364-374, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36103412

RESUMO

OBJECTIVE: This study was undertaken to determine the role of CD38, which can function as an enzyme to degrade NAD+ , in osteoarthritis (OA) development. METHODS: Human knee cartilage from normal donors and OA donors were examined for CD38 expression. "Gain-of-function," through overexpression of CD38 via transient transfection, and "loss-of-function," through pharmacologic inhibition of CD38, approaches were used to assess the effects of CD38 on intracellular NAD+ :NADH ratio and catabolic activity in chondrocytes. We also initiated joint injury-induced OA by surgical destabilization of the medial meniscus (DMM) in CD38 knockout mice and wild-type (WT; C57BL/6) mice and in WT male mice in the presence or absence of apigenin treatment. Cartilage degradation, synovial inflammation, subchondral bone changes, and pain behavior were evaluated after DMM surgery. We also examined expression of CD38 and the neuropeptide calcitonin gene-related peptide (CGRP) in knee sections from these mice. RESULTS: CD38 expression was up-regulated in human knee OA cartilage and in chondrocytes stimulated with the proinflammatory cytokine interleukin-1ß (IL-1ß). Overexpression of CD38 in chondrocytes resulted in reduced cellular NAD+ :NADH ratio and augmented catabolic responses to IL-1ß. These effects were reversed by pharmacologic inhibition of CD38. Cartilage degradation and synovial inflammation, associated with increased CD38 expression in cartilage and synovium, osteophyte formation and subchondral bone sclerosis, and pain-like behavior linked to increased CGRP expression in the synovium were observed in WT mice after joint injury. Such effects were significantly reduced in mice deficient in CD38 through either genetic knockout or pharmacologic inhibition. CONCLUSION: CD38 deficiency exerts OA disease-modifying effects. Inhibition of CD38 has the potential to be a novel therapeutic approach for OA treatment.


Assuntos
Cartilagem Articular , Osteoartrite do Joelho , Camundongos , Masculino , Humanos , Animais , NAD/metabolismo , NAD/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Cartilagem Articular/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Osteoartrite do Joelho/metabolismo , Inflamação/metabolismo , Dor/metabolismo , Camundongos Knockout
4.
Arthritis Res Ther ; 24(1): 175, 2022 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-35879786

RESUMO

OBJECTIVE: Linked metabolic and cardiovascular comorbidities are prevalent in hyperuricemia and gout. For mechanistic insight into impact on inflammatory processes and cardiometabolic risk factors of xanthine oxidase inhibitor urate-lowering therapy (ULT) titration to target, we performed a prospective study of gout serum metabolomes from a ULT trial. METHODS: Sera of gout patients meeting the 2015 ACR/EULAR gout classification criteria (n = 20) and with hyperuricemia were studied at time zero and weeks 12 and 24 of febuxostat or allopurinol dose titration ULT. Ultrahigh performance liquid chromatography-tandem mass spectroscopy acquired the serum spectra. Data were assessed using the Metabolon and Metaboloanalyst software. Lipolysis validation assays were done in febuxostat and/or colchicine-treated 3T3-L1 differentiated adipocytes. RESULTS: Serum urate decreased from time zero (8.21 ±1.139 SD) at weeks 12 (5.965 ± 1.734 SD) and 24 (5.655 ±1.763 SD). Top metabolites generated by changes in nucleotide and certain amino acid metabolism and polyamine pathways were enriched at 12 and 24 weeks ULT, respectively. Decreases in multiple fatty acid metabolites were observed at 24 weeks, linked with obesity. In cultured adipocytes, febuxostat significantly decreased while colchicine increased the lipolytic response to ß-adrenergic-agonism or TNF. CONCLUSION: Metabolomic profiles linked xanthine oxidase inhibitor-based ULT titration to target with reduced serum free fatty acids. In vitro validation studies revealed that febuxostat, but not colchicine, reduced lipolysis in cultured adipocytes. Since soluble urate, xanthine oxidase inhibitor treatment, and free fatty acids modulate inflammation, our findings suggest that by suppressing lipolysis, ULT could regulate inflammation in gout and comorbid metabolic and cardiovascular disease.


Assuntos
Gota , Hiperuricemia , Adipócitos , Alopurinol/uso terapêutico , Colchicina , Inibidores Enzimáticos , Ácidos Graxos não Esterificados/uso terapêutico , Febuxostat/farmacologia , Febuxostat/uso terapêutico , Supressores da Gota/efeitos adversos , Humanos , Inflamação/tratamento farmacológico , Lipólise , Estudos Prospectivos , Ácido Úrico , Xantina Oxidase/uso terapêutico
5.
Cell Rep ; 38(10): 110489, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263587

RESUMO

Monosodium urate crystals (MSUc) induce inflammation in vivo without prior priming, raising the possibility of an initial cell-autonomous phase. Here, using genome-wide transcriptomic analysis and biochemical assays, we demonstrate that MSUc alone induce a metabolic-inflammatory transcriptional program in non-primed human and murine macrophages that is markedly distinct to that induced by LPS. Genes uniquely upregulated in response to MSUc belong to lipid and amino acid metabolism, glycolysis, and SLC transporters. This upregulation leads to a metabolic rewiring in sera from individuals and mice with acute gouty arthritis. Mechanistically, the initiating inflammatory-metabolic changes in acute gout flares are regulated through a persistent expression and increased binding of JUN to the promoter of target genes through JNK signaling-but not P38-in a process that is different than after LPS stimulation and independent of inflammasome activation. Finally, pharmacological JNK inhibition limits MSUc-induced inflammation in animal models of acute gouty inflammation.


Assuntos
Artrite Gotosa , Ácido Úrico , Animais , Artrite Gotosa/induzido quimicamente , Artrite Gotosa/metabolismo , Inflamação/metabolismo , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Ácido Úrico/metabolismo
6.
Bioeng Transl Med ; 6(1): e10187, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33532587

RESUMO

Current drug delivery approaches for the treatment of cartilage disorders such as osteoarthritis (OA) remain inadequate to achieve sufficient drug penetration and retention in the dense cartilage matrix. Herein, we synthesize sub-30 nm lipid-polymer hybrid nanoparticles functionalized with collagen-targeting peptides for targeted drug delivery to the cartilage. The nanoparticles consist of a polymeric core for drug encapsulation and a lipid shell modified with a collagen-binding peptide. By combining these design features, the nanoparticles can penetrate deep and accumulate preferentially in the cartilage. Using MK-8722, an activator of 5'-adenosine monophosphate-activated protein kinase (AMPK), as a model drug, the nanoparticles can encapsulate the drug molecules in high capacity and release them in a sustained and controllable manner. When injected into the knee joints of the mice with collagenase-induced OA, the drug-loaded nanoparticles can effectively reduce cartilage damage and alleviate the disease severity. Overall, the ultrasmall targeted nanoparticles represent a promising delivery platform to overcome barriers of dense tissues for the treatment of various indications, including cartilage disorders.

7.
Methods Mol Biol ; 2245: 195-213, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33315204

RESUMO

Chondrocytes, the only cells in articular cartilage, are metabolically active and responsible for the turnover of extracellular matrix and maintenance of the tissue homeostasis. Changes in chondrocyte function can cause degradation of the matrix and loss of articular cartilage integrity, leading to development and progression of osteoarthritis (OA). These changes are exemplified by accumulated mitochondrial damage and dysfunction. Because mitochondria are the critical organelles to produce energy and play a key role in cellular processes, the approaches to assess mitochondrial function under both physiological and pathological conditions enable us to uncover the mechanisms on how dysfunction of mitochondria in chondrocytes mediates signaling pathways that are involved in disturbance of cartilage homeostasis. In this chapter, we describe the methods to evaluate mitochondrial biogenesis, activity and mitochondrial DNA (mtDNA) integrity in chondrocytes.


Assuntos
Condrócitos/metabolismo , DNA Mitocondrial/genética , DNA Mitocondrial/isolamento & purificação , Mitocôndrias/genética , Mitocôndrias/metabolismo , Biogênese de Organelas , Trifosfato de Adenosina , Animais , Biomarcadores , Cartilagem Articular/metabolismo , Regulação da Expressão Gênica , Humanos , Articulação do Joelho , Mutação , Osteoartrite/genética , Osteoartrite/metabolismo , Osteoartrite/patologia , Processamento de Proteína Pós-Traducional , Reação em Cadeia da Polimerase em Tempo Real
8.
Ann Rheum Dis ; 79(5): 635-645, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32156705

RESUMO

OBJECTIVES: In this study, we aim to determine the effect of metformin on osteoarthritis (OA) development and progression. METHODS: Destabilisation of the medial meniscus (DMM) surgery was performed in 10-week-old wild type and AMP-activated protein kinase (AMPK)α1 knockout (KO) mice. Metformin (4 mg/day in drinking water) was given, commencing either 2 weeks before or 2 weeks after DMM surgery. Mice were sacrificed 6 and 12 weeks after DMM surgery. OA phenotype was analysed by micro-computerised tomography (µCT), histology and pain-related behaviour tests. AMPKα1 (catalytic alpha subunit of AMPK) expression was examined by immunohistochemistry and immunofluorescence analyses. The OA phenotype was also determined by µCT and MRI in non-human primates. RESULTS: Metformin upregulated phosphorylated and total AMPK expression in articular cartilage tissue. Mild and more severe cartilage degeneration was observed at 6 and 12 weeks after DMM surgery, evidenced by markedly increased Osteoarthritis Research Society International scores, as well as reduced cartilage areas. The administration of metformin, commencing either before or after DMM surgery, caused significant reduction in cartilage degradation. Prominent synovial hyperplasia and osteophyte formation were observed at both 6 and 12 weeks after DMM surgery; these were significantly inhibited by treatment with metformin either before or after DMM surgery. The protective effects of metformin on OA development were not observed in AMPKα1 KO mice, suggesting that the chondroprotective effect of metformin is mediated by AMPK signalling. In addition, we demonstrated that treatment with metformin could also protect from OA progression in a partial medial meniscectomy animal model in non-human primates. CONCLUSIONS: The present study suggests that metformin, administered shortly after joint injury, can limit OA development and progression in injury-induced OA animal models.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Cartilagem Articular/efeitos dos fármacos , Metformina/farmacologia , Osteoartrite/tratamento farmacológico , Regulação para Cima/genética , Animais , Cartilagem Articular/patologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Hipoglicemiantes/farmacologia , Meniscos Tibiais/patologia , Meniscos Tibiais/cirurgia , Camundongos , Camundongos Knockout , Camundongos Obesos , Osteoartrite/patologia , Distribuição Aleatória , Sensibilidade e Especificidade , Transdução de Sinais/genética
9.
Arthritis Rheumatol ; 72(5): 802-814, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31738005

RESUMO

OBJECTIVE: In gout, autoinflammatory responses to urate crystals promote acute arthritis flares, but the pathogeneses of tophi, chronic synovitis, and erosion are less well understood. Defining the pathways of epigenomic immunity training can reveal novel pathogenetic factors and biomarkers. The present study was undertaken to seminally probe differential DNA methylation patterns utilizing epigenome-wide analyses in patients with gout. METHODS: Peripheral blood mononuclear cells (PBMCs) were obtained from a San Diego cohort of patients with gout (n = 16) and individually matched healthy controls (n = 14). PBMC methylome data were processed with ChAMP package in R. ENCODE data and Taiji data analysis software were used to analyze transcription factor (TF)-gene networks. As an independent validation cohort, whole blood DNA samples from New Zealand Maori subjects (n = 13 patients with gout, n = 16 control subjects without gout) were analyzed. RESULTS: Differentially methylated loci clearly separated gout patients from controls, as determined by hierarchical clustering and principal components analyses. IL23R, which mediates granuloma formation and cell invasion, was identified as one of the multiple differentially methylated gout risk genes. Epigenome-wide analyses revealed differential methylome pathway enrichment for B and T cell receptor signaling, Th17 cell differentiation and interleukin-17 signaling, convergent longevity regulation, circadian entrainment, and AMP-activated protein kinase signaling, which are pathways that impact inflammation via insulin-like growth factor 1 receptor, phosphatidylinositol 3-kinase/Akt, NF-κB, mechanistic target of rapamycin signaling, and autophagy. The gout cohorts overlapped for 37 (52.9%) of the 70 TFs with hypomethylated sequence enrichment and for 30 (78.9%) of the 38 enriched KEGG pathways identified via TFs. Evidence of shared differentially methylated gout TF-gene networks, including the NF-κB activation-limiting TFs MEF2C and NFATC2, pointed to osteoclast differentiation as the most strongly weighted differentially methylated pathway that overlapped in both gout cohorts. CONCLUSION: These findings of differential DNA methylation of networked signaling, transcriptional, innate and adaptive immunity, and osteoclastogenesis genes and pathways suggest that they could serve as novel therapeutic targets in the management of flares, tophi, chronic synovitis, and bone erosion in patients with gout.


Assuntos
Imunidade Adaptativa/genética , Metilação de DNA/fisiologia , Gota/genética , Gota/imunologia , Imunidade Inata/genética , Osteogênese/genética , Transdução de Sinais/genética , Transcrição Gênica , Estudos de Coortes , Feminino , Redes Reguladoras de Genes , Humanos , Leucócitos Mononucleares , Masculino
10.
Cell Metab ; 29(6): 1350-1362.e7, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-30982734

RESUMO

Choline is a vitamin-like nutrient that is taken up via specific transporters and metabolized by choline kinase, which converts it to phosphocholine needed for de novo synthesis of phosphatidylcholine (PC), the main phospholipid of cellular membranes. We found that Toll-like receptor (TLR) activation enhances choline uptake by macrophages and microglia through induction of the choline transporter CTL1. Inhibition of CTL1 expression or choline phosphorylation attenuated NLRP3 inflammasome activation and IL-1ß and IL-18 production in stimulated macrophages. Mechanistically, reduced choline uptake altered mitochondrial lipid profile, attenuated mitochondrial ATP synthesis, and activated the energy sensor AMP-activated protein kinase (AMPK). By potentiating mitochondrial recruitment of DRP1, AMPK stimulates mitophagy, which contributes to termination of NLRP3 inflammasome activation. Correspondingly, choline kinase inhibitors ameliorated acute and chronic models of IL-1ß-dependent inflammation.


Assuntos
Colina/metabolismo , Colina/farmacocinética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Animais , Butanos/farmacologia , Células Cultivadas , Síndromes Periódicas Associadas à Criopirina/genética , Síndromes Periódicas Associadas à Criopirina/metabolismo , Síndromes Periódicas Associadas à Criopirina/patologia , Feminino , Células HEK293 , Humanos , Absorção Intestinal/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Compostos de Piridínio/farmacologia
11.
Arthritis Res Ther ; 20(1): 204, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30189890

RESUMO

BACKGROUND: Arhalofenate acid, the active acid form of arhalofenate, is a non-agonist peroxisome proliferator-activated receptor γ (PPARγ) ligand, with uricosuric activity via URAT1 inhibition. Phase II studies revealed decreased acute arthritis flares in arhalofenate-treated gout compared with allopurinol alone. Hence, we investigated the anti-inflammatory effects and mechanisms of arhalofenate and its active acid form for responses to monosodium urate (MSU) crystals. METHODS: We assessed in-vivo responses to MSU crystals in murine subcutaneous air pouches and in-vitro responses in murine bone marrow-derived macrophages (BMDMs) by enzyme-linked immunosorbent assay (ELISA), SDS-PAGE/Western blot, immunostaining, and transmission electron microscopy analyses. RESULTS: Oral administration of arhalofenate (250 mg/kg) blunted total leukocyte ingress, neutrophil influx, and air pouch fluid interleukin (IL)-1ß, IL-6, and CXCL1 in response to MSU crystal injection (p < 0.05 for each). Arhalofenate acid (100 µM) attenuated MSU crystal-induced IL-1ß production in BMDMs via inhibition of NLRP3 inflammasome activation. In addition, arhalofenate acid dose-dependently increased activation (as assessed by phosphorylation) of AMP-activated protein kinase (AMPK). Studying AMPKα1 knockout mice, we elucidated that AMPK mediated the anti-inflammatory effects of arhalofenate acid. Moreover, arhalofenate acid attenuated the capacity of MSU crystals to suppress AMPK activity, regulated expression of multiple downstream AMPK targets that modulate mitochondrial function and oxidative stress, preserved intact mitochondrial cristae and volume density, and promoted anti-inflammatory autophagy flux in BMDMs. CONCLUSIONS: Arhalofenate acid is anti-inflammatory and acts via AMPK activation and its downstream signaling in macrophages. These effects likely contribute to a reduction of gout flares.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Acetamidas/farmacologia , Mediadores da Inflamação/metabolismo , Fenilacetatos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ácido Úrico/toxicidade , Acetamidas/uso terapêutico , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células Cultivadas , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Fenilacetatos/uso terapêutico , Transdução de Sinais/fisiologia , Ácido Úrico/antagonistas & inibidores
12.
Ann Rheum Dis ; 75(1): 286-94, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25362043

RESUMO

OBJECTIVE: AMP-activated protein kinase (AMPK) is metabolic biosensor with anti-inflammatory activities. Gout is commonly associated with excesses in soluble urate and in nutrition, both of which suppress tissue AMPK activity. Gout is driven by macrophage-mediated inflammation transduced partly by NLRP3 inflammasome activation and interleukin (IL)-1ß release. Hence, we tested the hypothesis that AMPK activation limits monosodium urate (MSU) crystal-induced inflammation. METHODS: We studied bone marrow-derived macrophages (BMDMs) from AMPKα1 knockout and wild-type mice, and assessed the selective AMPK pharmacological activator A-769662 and a low concentration (10 nM) of colchicine. We examined phosphorylation (activation) of AMPKα Thr172, NLRP3 mRNA expression, and caspase-1 cleavage and IL-1ß maturation using western blot and quantitative RT-PCR approaches. We also assessed subcutaneous murine air pouch inflammatory responses to MSU crystals in vivo. RESULTS: MSU crystals suppressed phosphorylation of AMPKα in BMDMs. Knockout of AMPKα1 enhanced, and, conversely, A-769662-inhibited MSU crystal-induced inflammatory responses including IL-1ß and CXCL1 release in vitro and in vivo. A-769662 promoted AMPK-dependent macrophage anti-inflammatory M2 polarisation and inhibited NLRP3 gene expression, activation of caspase-1 and IL-1ß. Colchicine, at low concentration (10 nM) achieved in gout flare prophylaxis dosing, promoted phosphorylation of AMPKα and macrophage M2 polarisation, and reduced activation of caspase-1 and release of IL-1ß and CXCL1 by MSU crystals in BMDMs in vitro. CONCLUSIONS: AMPK activity limits MSU crystal inflammation in vitro and in vivo, and transduces multiple anti-inflammatory effects of colchicine in macrophages. Targeting increased and sustained AMPK activation in inflammatory cells merits further investigation for enhancing efficacy of prophylaxis and treatment of gouty inflammation.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Colchicina/farmacologia , Supressores da Gota/farmacologia , Gota/enzimologia , Macrófagos/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/efeitos dos fármacos , Animais , Compostos de Bifenilo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Gota/induzido quimicamente , Gota/patologia , Macrófagos/fisiologia , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Pironas/farmacologia , Tiofenos/farmacologia , Ácido Úrico
13.
PLoS One ; 10(10): e0140452, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26474486

RESUMO

OBJECTIVE: AMP-activated protein kinase (AMPK) is a serine/threonine protein kinase critically involved in the regulation of cellular energy homeostasis. It is a central regulator of both lipid and glucose metabolism. Many studies have suggested that AMPK activation exert significant anti-inflammatory and immunosuppressive effects. In this study, we assessed whether targeted activation of AMPK inhibits inflammatory arthritis in vivo. METHODS: We tested the effect of A-769662, a specific AMPK agonist (60mg/kg/bid) in mouse models of antigen-induced arthritis (AIA) and passive K/BxN serum-induced arthritis. The passive K/BxN serum-induced arthritis model was also applied to AMPKα1-deficient mice. Joints were harvested and subjected to histological analysis. IL-6 expression was measured in both joint tissues and sera by ELISA. The effect of A-769662 on bone marrow derived macrophage (BMDM) response to stimulation with TLR2 and TLR4 agonists was tested in vitro. RESULTS: AMPK activation by A-769662 reduced inflammatory infiltration and joint damage in both mouse models. IL-6 expression in serum and arthritic joints was significantly decreased in A-769662-treated mice. AMPKα1 deficient mice mildly elicited an increase of clinical arthritis. IL-6 expression at both mRNA and protein levels, phosphorylation of p65 NF-κB and MAPK phosphorylation were inhibited by A-769662 in BMDMs stimulated with either TLR2 or TLR4 agonists. CONCLUSIONS: AMPK activation by specific AMPK agonist A-769662 suppressed inflammatory arthritis in mice as well as IL-6 expression in serum and arthritic joints. These data suggest that targeted activation of AMPK has a potential to be an effective therapeutic strategy for IL-6 dependent inflammatory arthritis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-6/metabolismo , Pironas/farmacologia , Tiofenos/farmacologia , Proteínas Quinases Ativadas por AMP/deficiência , Animais , Artrite Experimental/enzimologia , Artrite Experimental/imunologia , Compostos de Bifenilo , Células da Medula Óssea/citologia , Ativação Enzimática/efeitos dos fármacos , Interleucina-6/genética , Articulações/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pironas/uso terapêutico , Tiofenos/uso terapêutico
14.
Arthritis Rheumatol ; 66(11): 3073-82, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25047750

RESUMO

OBJECTIVE: AMP-activated protein kinase (AMPK) inhibits chondrocyte procatabolic responses to inflammation and biomechanical injury. This study was undertaken to test the hypothesis that peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and FoxO3A, 2 major AMPK downstream targets, mediate the chondroprotective effect of AMPK activation. METHODS: We assessed the activity of AMPKα (threonine 172 phosphorylation) and the expression of PGC-1α and FoxO3A in human chondrocytes and AMPKα1- or AMPKα2-knockout mouse chondrocytes by Western blotting, and in mouse knee cartilage by immunohistochemistry. We also knocked down or overexpressed PGC-1α and FoxO3A by small interfering RNA or plasmid DNA transfection, respectively. We assessed mitochondrial superoxide generation using MitoSOX Red. RESULTS: Expression of PGC-1α and FoxO3A was enhanced by pharmacologic AMPK activator A-769662 but impaired in AMPKα1(-/-) or AMPKα2(-/-) mouse chondrocytes. Reduced expression of PGC-1α and FoxO3A was observed in mouse knee instability-induced osteoarthritis (OA) cartilage and in aged C57BL/6 mouse knee cartilage. Knockdown of PGC-1α and FoxO3A enhanced, but limited the ability of A-769662 to inhibit, phosphorylation of p65 NF-κB (Ser(536) ) and procatabolic responses induced by inflammatory cytokines. Forced expression of PGC-1α and FoxO3A induced increased expression of superoxide dismutase 2 (SOD2) and catalase, but A-769662 failed to increase the expression of SOD2 and catalase in either PGC-1α- or FoxO3A-knockdown chondrocytes. Last, menadione-induced superoxide generation was inhibited by AMPK pharmacologic activators and by overexpression of PGC-1α or FoxO3A. CONCLUSION: PGC-1α and FoxO3A limit oxidative stress and at least partially mediate the capacity of AMPK activity to block procatabolic responses in chondrocytes, and therefore have the potential to inhibit the progression of cartilage damage in OA.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Condrócitos/metabolismo , Condrócitos/patologia , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Quinases Ativadas por AMP/deficiência , Proteínas Quinases Ativadas por AMP/genética , Envelhecimento/metabolismo , Animais , Catalase/metabolismo , Células Cultivadas , Condrócitos/efeitos dos fármacos , Modelos Animais de Doenças , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Humanos , Meniscos Tibiais/cirurgia , Camundongos , Camundongos Endogâmicos C57BL , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Estresse Oxidativo/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , RNA Interferente Pequeno/farmacologia , Superóxido Dismutase/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção
15.
Atherosclerosis ; 222(2): 417-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22552116

RESUMO

OBJECTIVE: Intima formation involves smooth muscle cell (SMC) proliferation and migration that ultimately drives arterial stenosis, thrombosis, and ischemia in atherosclerosis, hypertension, and arterial revascularization. Receptor for advanced glycation endproducts (RAGE) is a transmembrane signaling receptor implicated in diabetic renal and vascular complications, and post-injury intima formation, partly via Signal transducer and activator of transcription 3 (STAT3) activation. The metabolic super-regulator Adenosine monophosphate kinase (AMPK) inhibits SMC proliferation and intima formation. AMPK activation is promoted by liver kinase B1 (LKB1), and LKB1 inhibits STAT3 activation. Here, we tested the hypothesis that RAGE promotes arterial intima formation by modulating both LKB1 and AMPK. METHODS AND RESULTS: RAGE ligands (the calgranulin S100A11, and glycated albumin) suppressed AMPK activation in conjunction with increased proliferation and migration of cultured SMCs. These effects were inhibited both by RAGE deficiency and by prior AMPK activation. In SMCs, RAGE ligands decreased LKB1 activity. Moreover, knockdown of both LKB1 and AMPK were associated with increased STAT3 phosphorylation levels. In response to murine carotid artery ligation, expression of RAGE and S100A11 increased, whereas AMPK and LKB1 activity decreased in situ. Conversely, LKB1 and AMPK activity increased in situ, and neointima formation was attenuated in Rage(-/-) mice. CONCLUSION: The linkage of decreased LKB1 and AMPK activity with increased STAT3 in SMCs mediates the capacity of RAGE ligand-induced signaling to promote neointima formation in response to arterial injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Lesões das Artérias Carótidas/enzimologia , Fígado/enzimologia , Músculo Liso Vascular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Ativação Enzimática , Técnicas de Silenciamento de Genes , Produtos Finais de Glicação Avançada , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Neointima , Fosforilação , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/genética , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Proteínas S100/metabolismo , Fator de Transcrição STAT3/metabolismo , Albumina Sérica/metabolismo , Transdução de Sinais , Albumina Sérica Glicada
16.
Arthritis Rheum ; 63(7): 1928-37, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21400477

RESUMO

OBJECTIVE: Interleukin-1ß (IL-1ß) and tumor necrosis factor α (TNFα) stimulate chondrocyte matrix catabolic responses, thereby compromising cartilage homeostasis in osteoarthritis (OA). AMP-activated protein kinase (AMPK), which regulates energy homeostasis and cellular metabolism, also exerts antiinflammatory effects in multiple tissues. This study was undertaken to test the hypothesis that AMPK activity limits chondrocyte matrix catabolic responses to IL-1ß and TNFα. METHODS: Expression of AMPK subunits was examined, and AMPKα activity was ascertained by the phosphorylation status of AMPKα Thr(172) in human knee articular chondrocytes and cartilage by Western blotting and immunohistochemistry, respectively. Procatabolic responses to IL-1ß and TNFα, such as release of glycosaminoglycan, nitric oxide, and matrix metalloproteinases 3 and 13 were determined by dimethylmethylene blue assay, Griess reaction, and Western blotting, respectively, in cartilage explants and chondrocytes with and without knockdown of AMPKα by small interfering RNA. RESULTS: Normal human knee articular chondrocytes expressed AMPKα1, α2, ß1, ß2, and γ1 subunits. AMPK activity was constitutively present in normal articular chondrocytes and cartilage, but decreased in OA articular chondrocytes and cartilage and in normal chondrocytes treated with IL-1ß and TNFα. Knockdown of AMPKα resulted in enhanced catabolic responses to IL-1ß and TNFα in chondrocytes. Moreover, AMPK activators suppressed cartilage/chondrocyte procatabolic responses to IL-1ß and TNFα and the capacity of TNFα and CXCL8 (IL-8) to induce type X collagen expression. CONCLUSION: Our findings indicate that AMPK activity is reduced in OA cartilage and in chondrocytes following treatment with IL-1ß or TNFα. AMPK activators attenuate dephosphorylation of AMPKα and procatabolic responses in chondrocytes induced by these cytokines. These observations suggest that maintenance of AMPK activity supports cartilage homeostasis by protecting cartilage matrix from inflammation-induced degradation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Condrócitos/metabolismo , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Western Blotting , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Condrócitos/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Interleucina-1beta/farmacologia , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/metabolismo , Metaloproteinases da Matriz/metabolismo , Osteoartrite/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
17.
Arthritis Rheum ; 62(7): 2170-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20506351

RESUMO

OBJECTIVE: The mechanism by which monosodium urate monohydrate (MSU) crystals intracellularly activate the cryopyrin inflammasome is unknown. The aim of this study was to use a mouse molecular genetics-based approach to test whether the leucine-rich repeat (LRR) domain of cryopyrin is required for MSU crystal-induced inflammation. METHODS: Cryopyrin-knockout lacZ (Cryo(-Z/-Z)) mice and mice with the cryopyrin LRR domain deleted and fused to the lacZ reporter (Cryo(DeltaLRR Z/DeltaLRR Z)) were generated using bacterial artificial chromosome-based targeting vectors, which allow for large genomic deletions. Bone marrow-derived macrophages from Cryo(DeltaLRR Z/DeltaLRR Z) mice, Cryo(-Z/-Z) mice, and congenic wild-type (WT) mice were challenged with endotoxin-free MSU crystals under serum-free conditions. Phagocytosis and cytokine expression were assessed by flow cytometry and enzyme-linked immunosorbent assay. MSU crystals also were injected into mouse synovial-like subcutaneous air pouches. The in vivo inflammatory responses were examined. RESULTS: Release of interleukin-1beta (IL-1beta), but not CXCL1 and tumor necrosis factor alpha, was impaired in Cryo(DeltaLRR Z/DeltaLRR Z) and Cryo(-Z/-Z) mouse bone marrow-derived macrophages compared with WT mouse bone marrow-derived macrophages in response to not only MSU crystals but also other known stimuli that activate the cryopyrin inflammasome. In addition, a comparable percentage of MSU crystals taken up by each type of bone marrow-derived macrophage was observed. Moreover, total leukocyte infiltration in the air pouch and IL-1beta production were attenuated in Cryo(-Z/-Z) and Cryo(DeltaLRR Z/DeltaLRR Z) mice at 6 hours postinjection of MSU crystals compared with WT mice. CONCLUSION: MSU crystal-induced inflammatory responses were comparably attenuated both in vitro and in vivo in Cryo(DeltaLRR Z/DeltaLRR Z) and Cryo(-Z/-Z) mice. Hence, the LRR domain of cryopyrin plays a role in mediating MSU crystal-induced inflammation in this model.


Assuntos
Artrite Gotosa/metabolismo , Proteínas de Transporte/metabolismo , Proteínas/metabolismo , Sequências Repetitivas de Aminoácidos , Animais , Artrite Gotosa/imunologia , Células da Medula Óssea , Proteínas de Transporte/imunologia , Células Cultivadas , Citocinas/metabolismo , Feminino , Proteínas de Repetições Ricas em Leucina , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fagocitose/efeitos dos fármacos , Proteínas/genética , Proteínas/imunologia , Pele/efeitos dos fármacos , Pele/patologia , Ácido Úrico/imunologia , Ácido Úrico/toxicidade
18.
Arthritis Rheum ; 60(12): 3642-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19950258

RESUMO

OBJECTIVE: Interleukin-1beta (IL-1beta) is a key cytokine linked to the pathogenesis of acute arthritis. Caspase 1, neutrophil elastase, and chymase all process proIL-1beta to its biologically active form. This study was undertaken to examine the potential contributions of each of these proteases in experimental models of inflammatory arthritis. METHODS: Caspase 1-deficient (Casp1-/-) and wild-type (WT) mice were tested for their response to arthritogenic K/BxN serum transfer for induction of arthritis or injection of monosodium urate monohydrate (MSU) crystals for induction of peritonitis. All mice were prophylactically treated with inhibitors of neutrophil elastase or chymase. Arthritic paws were tested for the presence of IL-1beta protein by enzyme-linked immunosorbent assay and Western blotting. Neutrophils and mast cells from WT and mutant mice were tested for their ability to secrete IL-1beta after in vitro stimulation, in the presence of protease inhibitors. RESULTS: Casp1-/- and WT mice developed paw swelling to the same extent in the K/BxN serum transfer-induced arthritis model. MSU crystal injection into Casp1-/- mice also resulted in neutrophil influx and production of measurable peritoneal IL-1beta protein. Both of these responses were attenuated with neutrophil elastase inhibitors. K/BxN serum transfer-induced arthritis was also reduced by treatment with a chymase inhibitor. Casp1-/- neutrophils and mast cells, when exposed to MSU crystals, secreted similar amounts of IL-1beta protein upon in vitro stimulation with lipopolysaccharide, albeit at lower levels than that secreted by WT cells. Elastase and chymase inhibitors reduced the amount of IL-1beta released by these cells. CONCLUSION: The production of IL-1beta by neutrophils and mast cells is not exclusively dependent on caspase 1, and other proteases can compensate for the loss of caspase 1 in vivo. These pathways might therefore compromise the caspase 1-targeted therapies in neutrophil-predominant arthritis.


Assuntos
Artrite Experimental/enzimologia , Caspase 1/deficiência , Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Peritonite/enzimologia , Precursores de Proteínas/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Autoanticorpos/imunologia , Caspase 1/genética , Caspase 1/metabolismo , Células Cultivadas , Quimases/metabolismo , Edema , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Inativação Gênica , Membro Posterior , Imunização Passiva , Articulações/imunologia , Articulações/metabolismo , Articulações/patologia , Elastase de Leucócito/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , Mastócitos/efeitos dos fármacos , Mastócitos/enzimologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mieloblastina/metabolismo , Infiltração de Neutrófilos , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Neutrófilos/imunologia , Peritonite/imunologia , Peritonite/fisiopatologia
19.
J Biol Chem ; 282(48): 35279-92, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17916553

RESUMO

Obesity and type 2 diabetes are characterized by decreased insulin sensitivity, elevated concentrations of free fatty acids (FFAs), and increased macrophage infiltration in adipose tissue (AT). Here, we show that FFAs can cause activation of RAW264.7 cells primarily via the JNK signaling cascade and that TLR2 and TLR4 are upstream of JNK and help transduce FFA proinflammatory signals. We also demonstrate that F4/80(+)CD11b(+)CD11c(+) bone marrow-derived dendritic cells (BMDCs) have heightened proinflammatory activity compared with F4/80(+)CD11b(+)CD11c(-) bone marrow-derived macrophages and that the proinflammatory activity and JNK phosphorylation of BMDCs, but not bone marrow-derived macrophages, was further increased by FFA treatment. F4/80(+)CD11b(+)CD11c(+) cells were found in AT, and the proportion and number of these cells in AT is increased in ob/ob mice and by feeding wild type mice a high fat diet for 1 and 12 weeks. AT F4/80(+)CD11b(+)CD11c(+) cells express increased inflammatory markers compared with F4/80(+)CD11b(+)CD11c(-) cells, and FFA treatment increased inflammatory responses in these cells. In addition, we found that CD11c expression is increased in skeletal muscle of high fat diet-fed mice and that conditioned medium from FFA-treated wild type BMDCs, but not TLR2/4 DKO BMDCs, can induce insulin resistance in L6 myotubes. Together our results show that FFAs can activate CD11c(+) myeloid proinflammatory cells via TLR2/4 and JNK signaling pathways, thereby promoting inflammation and subsequent cellular insulin resistance.


Assuntos
Tecido Adiposo/metabolismo , Ácidos Graxos não Esterificados/metabolismo , MAP Quinase Quinase 4/metabolismo , Macrófagos/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células da Medula Óssea/metabolismo , Antígeno CD11b/biossíntese , Antígeno CD11c/biossíntese , Inflamação , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transdução de Sinais
20.
J Immunol ; 177(9): 6370-8, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17056568

RESUMO

Phagocyte ingestion of monosodium urate (MSU) crystals can induce proinflammatory responses and trigger acute gouty inflammation. Alternatively, the uptake of MSU crystals by mature macrophages can be noninflammatory and promote resolution of gouty inflammation. Macrophage activation by extracellular MSU crystals involves apparent recognition and ingestion mediated by TLR2 and TLR4, with subsequent intracellular recognition linked to caspase-1 activation and IL-1beta processing driven by the NACHT-LRR-PYD-containing protein-3 inflammasome. In this study, we examined the potential role in gouty inflammation of CD14, a phagocyte-expressed pattern recognition receptor that functionally interacts with both TLR2 and TLR4. MSU crystals, but not latex beads, directly bound recombinant soluble (s) CD14 in vitro. CD14(-/-) bone marrow-derived macrophages (BMDMs) demonstrated unimpaired phagocytosis of MSU crystals but reduced p38 phosphorylation and approximately 90% less IL-1beta and CXCL1 release. Attenuated MSU crystal-induced IL-1beta release in CD14(-/-) BMDMs was mediated by decreased pro-IL-1beta protein expression and additionally by decreased caspase-1 activation and IL-1beta processing consistent with diminished NACHT-LRR-PYD-containing protein-3 inflammasome activation. Coating of MSU crystals with sCD14, but not sTLR2 or sTLR4, restored IL-1beta and CXCL1 production in CD14(-/-) BMDMs in vitro. Gain of function of CD14 directly enhanced TLR4-mediated signaling in response to MSU crystals in transfected Chinese hamster ovary cells in vitro. Last, MSU crystal-induced leukocyte influx at 6 h was reduced by approximately 75%, and local induction of IL-1beta decreased by >80% in CD14(-/-) mouse s.c. air pouches in vivo. We conclude that engagement of CD14 is a central determinant of the inflammatory potential of MSU crystals.


Assuntos
Gota/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Fagocitose , Ácido Úrico/metabolismo , Animais , Células CHO , Caspase 1/metabolismo , Quimiocinas CXC/metabolismo , Cricetinae , Cristalização , Ativação Enzimática , Gota/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Receptores de Lipopolissacarídeos/genética , Macrófagos/efeitos dos fármacos , Camundongos , Fagocitose/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptor 4 Toll-Like/metabolismo , Ácido Úrico/química , Ácido Úrico/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA