Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Inflamm Res ; 73(5): 739-751, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38493256

RESUMEN

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.


Asunto(s)
ADP-Ribosil Ciclasa 1 , Inflamación , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , NAD , Ácido Úrico , Animales , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Inflamación/tratamiento farmacológico , Ratones , NAD/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Células Cultivadas , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/metabolismo , Artritis Gotosa/genética , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos
2.
Ann Rheum Dis ; 79(5): 635-645, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32156705

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Cartílago Articular/efectos de los fármacos , Metformina/farmacología , Osteoartritis/tratamiento farmacológico , Regulación hacia Arriba/genética , Animales , Cartílago Articular/patología , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Hipoglucemiantes/farmacología , Meniscos Tibiales/patología , Meniscos Tibiales/cirugía , Ratones , Ratones Noqueados , Ratones Obesos , Osteoartritis/patología , Distribución Aleatoria , Sensibilidad y Especificidad , Transducción de Señal/genética
3.
J Biol Chem ; 293(31): 12259-12270, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-29929979

RESUMEN

Certain dysregulated chondrocyte metabolic adaptive responses such as decreased activity of the master regulator of energy metabolism AMP-activated protein kinase (AMPK) promote osteoarthritis (OA). Metabolism intersects with epigenetic and transcriptional responses. Hence, we studied chondrocyte ATP-citrate lyase (ACLY), which generates acetyl-CoA from mitochondrial-derived citrate, and modulates acetylation of histones and transcription factors. We assessed ACLY in normal and OA human knee chondrocytes and cartilages by Western blotting and immunohistochemistry, and quantified acetyl-CoA fluorometrically. We examined histone and transcription factor lysine acetylation by Western blotting, and assessed histone H3K9 and H3K27 occupancy of iNOS, MMP3, and MMP13 promoters by chromatin immunoprecipitation (ChIP) and quantitative PCR (qPCR). We analyzed iNOS, MMP3, MMP13, aggrecan (ACAN), and Col2a1 gene expression by RT-qPCR. Glucose availability regulated ACLY expression and function, nucleocytosolic acetyl-CoA, and histone acetylation. Human knee OA chondrocytes exhibited increased ACLY activation (assessed by Ser-455 phosphorylation), associated with increased H3K9 and H3K27 acetylation. Inhibition of ACLY attenuated IL-1ß-induced transcription of iNOS, MMP3, and MMP13 by suppressing acetylation of p65 NF-κB, H3K9, and H3K27, blunted release of NO, MMP3, and MMP13, and also reduced SOX9 acetylation that promoted SOX9 nuclear translocation, leading to increased aggrecan and Col2a1 mRNA expression. ACLY is a novel player involved in regulation of cartilage matrix metabolism. Increased ACLY activity in OA chondrocytes increased nucleocytosolic acetyl-CoA, leading to increased matrix catabolism via dysregulated histone and transcription factor acetylation. Pharmacologic ACLY inhibition in OA chondrocytes globally reverses these changes and stimulates matrix gene expression and AMPK activation, supporting translational investigation in OA.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Cartílago Articular/enzimología , Condrocitos/enzimología , Matriz Extracelular/enzimología , Osteoartritis de la Rodilla/enzimología , ATP Citrato (pro-S)-Liasa/genética , Acetilcoenzima A/metabolismo , Acetilación , Agrecanos/genética , Agrecanos/metabolismo , Cartílago Articular/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Histonas/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/metabolismo
4.
Ann Rheum Dis ; 75(1): 286-94, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25362043

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Colchicina/farmacología , Supresores de la Gota/farmacología , Gota/enzimología , Macrófagos/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/deficiencia , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Animales , Compuestos de Bifenilo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Gota/inducido químicamente , Gota/patología , Macrófagos/fisiología , Ratones Noqueados , Fosforilación/efectos de los fármacos , Pironas/farmacología , Tiofenos/farmacología , Ácido Úrico
5.
J Immunol ; 191(4): 1865-72, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23842751

RESUMEN

We have previously shown that i.m. administration of bacterially expressed murine histidyl-tRNA synthetase (HRS) triggers florid muscle inflammation (relative to appropriate control proteins) in various congenic strains of mice. Because severe disease develops even in the absence of adaptive immune responses to HRS, we sought to identify innate immune signaling components contributing to our model of HRS-induced myositis. In vitro stimulation assays demonstrated HRS-mediated activation of HEK293 cells transfected with either TLR2 or TLR4, revealing an excitatory capacity exceeding that of other bacterially expressed fusion proteins. Corresponding to this apparent functional redundancy of TLR signaling pathways, HRS immunization of B6.TLR2(-/-) and B6.TLR4(-/-) single-knockout mice yielded significant lymphocytic infiltration of muscle tissue comparable to that produced in C57BL/6 wild-type mice. In contrast, concomitant elimination of TLR2 and TLR4 signaling in B6.TLR2(-/-).TLR4(-/-) double-knockout mice markedly reduced the severity of HRS-induced muscle inflammation. Complementary subfragment analysis demonstrated that aa 60-90 of HRS were absolutely required for in vitro as well as in vivo signaling via these MyD88-dependent TLR pathways--effects mediated, in part, through preferential binding of exogenous ligands capable of activating specific TLRs. Collectively, these experiments indicate that multiple MyD88-dependent signaling cascades contribute to this model of HRS-induced myositis, underscoring the antigenic versatility of HRS and confirming the importance of innate immunity in this system.


Asunto(s)
Autoantígenos/toxicidad , Histidina-ARNt Ligasa/toxicidad , Factor 88 de Diferenciación Mieloide/fisiología , Enfermedad Autoinmune Experimental del Sistema Nervioso/fisiopatología , Transducción de Señal/fisiología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Animales , Autoantígenos/química , Autoantígenos/inmunología , Diglicéridos/metabolismo , Endotoxinas/metabolismo , Femenino , Proteína HMGB1/metabolismo , Histidina-ARNt Ligasa/química , Histidina-ARNt Ligasa/inmunología , Inmunidad Innata , Inmunización , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/toxicidad , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Enfermedad Autoinmune Experimental del Sistema Nervioso/etiología , Oligopéptidos/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Unión Proteica , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/efectos de los fármacos , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/efectos de los fármacos
6.
STAR Protoc ; 5(2): 103030, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38678566

RESUMEN

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.


Asunto(s)
Cristalización , Gota , Macrófagos , Ácido Úrico , Ácido Úrico/metabolismo , Ácido Úrico/química , Animales , Humanos , Ratones , Macrófagos/metabolismo , Gota/metabolismo
7.
Res Sq ; 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38260556

RESUMEN

Background: Effective xanthine oxidoreductase inhibition (XOI) urate-lowering treatment (ULT) to target significantly reduces gout flare burden and synovitis between 1-2 years therapy, without clearing all monosodium urate crystal deposits. Paradoxically, treat to target ULT is associated with increased flare activity for at least 1 year in duration on average, before gout flare burden decreases. Since XOI has anti-inflammatory effects, we tested for biomarkers of sustained, effective ULT that alters gouty inflammation. Methods: We characterized the proteome of febuxostat-treated murine bone marrow macrophages. Blood samples (baseline and 48 weeks ULT) were analyzed by unbiased proteomics in febuxostat and allopurinol ULT responders from two, independent, racially and ethnically distinct comparative effectiveness trial cohorts (n=19, n=30). STRING-db and multivariate analyses supplemented determinations of significantly altered proteins via Wilcoxon matched pairs signed rank testing. Results: The proteome of cultured IL-1b-stimulated macrophages revealed febuxostat-induced anti-inflammatory changes, including for classical and alternative pathway complement activation pathways. At 48 weeks ULT, with altered purine metabolism confirmed by serum metabolomics, serum urate dropped >30%, to normal (<6.8 mg/dL) in all the studied patients. Overall, flares declined from baseline. Treated gout patient sera and peripheral blood mononuclear cells (PBMCs) showed significantly altered proteins (p<0.05) in clustering and proteome networks. CRP was not a useful therapy response biomarker. By comparison, significant serum proteome changes included decreased complement C8 heterotrimer C8A and C8G chains essential for C5b-9 membrane attack complex assembly and function; increase in the NLRP3 inflammasome activation promoter vimentin; increased urate crystal phagocytosis inhibitor sCD44; increased gouty inflammation pro-resolving mediator TGFB1; decreased phagocyte-recruiting chemokine PPBP/CXCL7, and increased monocyte/macrophage-expressed keratin-related proteins (KRT9,14,16) further validated by PBMC proteomics. STRING-db analyses of significantly altered serum proteins from both cohorts revealed a tight interactome network including central mediators of gouty inflammation (eg, IL-1B, CXCL8, IL6, C5). Conclusions: Rewiring of inflammation mediators in a tight serum protein interactome was a biomarker of sustained XOI-based ULT that effectively reduced serum urate and gout flares. Monitoring of the serum and PBMC proteome, including for changes in the complement pathway could help determine onset and targets of anti-inflammatory changes in response to effective, sustained XOI-based ULT.Trial Registration: ClinicalTrials.gov Identifier: NCT02579096.

8.
Res Sq ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38766125

RESUMEN

Background: Urate-lowering treatment (ULT) to target with xanthine oxidase inhibitors (XOIs) paradoxically causes early increase in gouty arthritis flares. Because delayed reduction in flare burden is mechanistically unclear, we tested for ULT inflammation responsiveness markers. Methods: Unbiased proteomics analyzed blood samples (baseline, 48 weeks ULT) in two, independent ULT out trial cohorts (n = 19, n = 30). STRING-db and multivariate analyses supplemented determinations of altered proteins via Wilcoxon matched pairs signed rank testing in XOI ULT responders. Mechanistic studies characterized proteomes of cultured XOI-treated murine bone marrow macrophages (BMDMs). Results: At 48 weeks ULT, serum urate normalized in all gout patients, and flares declined, with significantly altered proteins (p < 0.05) in clustering and proteome networks in sera and peripheral blood mononuclear cells. Serum proteome changes included decreased complement C8 heterotrimer C8A and C8G chains and chemokine PPBP/CXCL7, and increased urate crystal phagocytosis inhibitor sCD44. In both cohorts, a treatment-emergent serum interactome included key gouty inflammation mediators (C5, IL-1B, CXCL8, IL6). Last, febuxostat inhibited complement activation pathway proteins in cultured BMDMs. Conclusions: Reduced gout flares are kinked with a XOI-treatment emergent complement- and inflammation-regulatory serum protein interactome. Serum and leukocyte proteomes could help identify onset of anti-inflammatory responsiveness to ULT in gout. Trial registration: ClinicalTrials.gov Identifier: NCT02579096, posted October 19, 2015.

9.
Curr Rheumatol Rep ; 15(5): 323, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23516014

RESUMEN

This review focuses on the recent advancements in the understanding of innate immunity in the pathogenesis of osteoarthritis, particularly with attention to the roles of damage-associated molecular patterns (DAMPs), pattern recognition receptors (PPRs), and complement in synovitis development and cartilage degradation. Endogenous molecular products derived from cellular stress and extracellular matrix disruption can function as DAMPs to induce inflammatory responses and pro-catabolic events in vitro and promote synovitis and cartilage degradation in vivo via PRRs. Some of the DAMPs and PRRs display various capacities in driving synovitis and/or cartilage degradation in different models of animal studies. New findings reveal that the inflammatory complement cascade plays a key in the pathogenesis of OA. Crosstalk between joint tissues such as synovium and cartilage communicated at the cellular level within the innate immune inflammatory network is implicated to play an important role in OA progression. Further studies on how the innate immune inflammatory network impacts the OA disease process at different stages of progression will lead to the development of new therapeutic strategies.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Osteoartritis/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Membrana Sinovial/inmunología , Sinovitis/inmunología , Animales , Apoptosis/inmunología , Cartílago Articular/inmunología , Cartílago Articular/patología , Progresión de la Enfermedad , Humanos , Osteoartritis/patología , Receptor Cross-Talk , Transducción de Señal , Membrana Sinovial/patología , Sinovitis/patología
10.
Arthritis Rheumatol ; 75(3): 364-374, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36103412

RESUMEN

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.


Asunto(s)
Cartílago Articular , Osteoartritis de la Rodilla , Ratones , Masculino , Humanos , Animales , NAD/metabolismo , NAD/farmacología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/farmacología , Cartílago Articular/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Osteoartritis de la Rodilla/metabolismo , Inflamación/metabolismo , Dolor/metabolismo , Ratones Noqueados
11.
Arthritis Rheumatol ; 75(5): 794-805, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36457235

RESUMEN

OBJECTIVE: In gout, hyperuricemia promotes urate crystal deposition, which stimulates the NLRP3 inflammasome and interleukin-1ß (IL-1ß)-mediated arthritis. Incident gout without background hyperuricemia is rarely reported. To identify hyperuricemia-independent mechanisms driving gout incidence and progression, we characterized erosive urate crystalline inflammatory arthritis in a young female patient with normouricemia diagnosed as having sufficient and weighted classification criteria for gout according to the American College of Rheumatology (ACR)/EULAR gout classification criteria (the proband). METHODS: We conducted whole-genome sequencing, quantitative proteomics, whole-blood RNA-sequencing analysis using serum samples from the proband. We used a mouse model of IL-1ß-induced knee synovitis to characterize proband candidate genes, biomarkers, and pathogenic mechanisms of gout. RESULTS: Lubricin level was attenuated in human proband serum and associated with elevated acute-phase reactants and inflammatory whole-blood transcripts and transcriptional pathways. The proband had predicted damaging gene variants of NLRP3 and of inter-α trypsin inhibitor heavy chain 3, an inhibitor of lubricin-degrading cathepsin G. Changes in the proband's serum protein interactome network supported enhanced lubricin degradation, with cathepsin G activity increased relative to its inhibitors, SERPINB6 and thrombospondin 1. Activation of Toll-like receptor 2 (TLR-2) suppressed levels of lubricin mRNA and lubricin release in cultured human synovial fibroblasts (P < 0.01). Lubricin blunted urate crystal precipitation and IL-1ß induction of xanthine oxidase and urate in cultured macrophages (P < 0.001). In lubricin-deficient mice, injection of IL-1ß in knees increased xanthine oxidase-positive synovial resident M1 macrophages (P < 0.05). CONCLUSION: Our findings linked normouricemic erosive gout to attenuated lubricin, with impaired control of cathepsin G activity, compounded by deleterious NLRP3 variants. Lubricin suppressed monosodium urate crystallization and blunted IL-1ß-induced increases in xanthine oxidase and urate in macrophages. The collective activities of articular lubricin that could limit incident and erosive gouty arthritis independently of hyperuricemia are subject to disruption by inflammation, activated cathepsin G, and synovial fibroblast TLR-2 signaling.


Asunto(s)
Artritis Gotosa , Gota , Hiperuricemia , Femenino , Humanos , Ratones , Animales , Receptor Toll-Like 2/genética , Catepsina G/efectos adversos , Ácido Úrico , Proteína con Dominio Pirina 3 de la Familia NLR , Xantina Oxidasa , Gota/genética , Inflamación/metabolismo , Interleucina-1beta/metabolismo
12.
Arthritis Rheum ; 63(7): 1928-37, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21400477

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Condrocitos/metabolismo , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Western Blotting , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos/efectos de los fármacos , Humanos , Inmunohistoquímica , Interleucina-1beta/farmacología , Articulación de la Rodilla/efectos de los fármacos , Articulación de la Rodilla/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Osteoartritis/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
13.
Cell Rep ; 38(10): 110489, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263587

RESUMEN

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.


Asunto(s)
Artritis Gotosa , Ácido Úrico , Animales , Artritis Gotosa/inducido químicamente , Artritis Gotosa/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ácido Úrico/metabolismo
14.
Arthritis Res Ther ; 24(1): 175, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35879786

RESUMEN

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.


Asunto(s)
Gota , Hiperuricemia , Adipocitos , Alopurinol/uso terapéutico , Colchicina , Inhibidores Enzimáticos , Ácidos Grasos no Esterificados/uso terapéutico , Febuxostat/farmacología , Febuxostat/uso terapéutico , Supresores de la Gota/efectos adversos , Humanos , Inflamación/tratamiento farmacológico , Lipólisis , Estudios Prospectivos , Ácido Úrico , Xantina Oxidasa/uso terapéutico
15.
Geroscience ; 44(4): 2195-2211, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35381951

RESUMEN

Elevated serum urate (hyperuricemia) promotes crystalline monosodium urate tissue deposits and gout, with associated inflammation and increased mortality. To identify modifiers of uric acid pathologies, we performed a fly Genome-Wide Association Study (GWAS) on purine metabolites using the Drosophila Genetic Reference Panel strains. We tested the candidate genes using the Drosophila melanogaster model of hyperuricemia and uric acid crystallization ("concretion formation") in the kidney-like Malpighian tubule. Medusa (mda) activity increased urate levels and inflammatory response programming. Conversely, whole-body mda knockdown decreased purine synthesis precursor phosphoribosyl pyrophosphate, uric acid, and guanosine levels; limited formation of aggregated uric acid concretions; and was sufficient to rescue lifespan reduction in the fly hyperuricemia and gout model. Levels of mda homolog FAM214A were elevated in inflammatory M1- and reduced in anti-inflammatory M2-differentiated mouse bone marrow macrophages, and influenced intracellular uric acid levels in human HepG2 transformed hepatocytes. In conclusion, mda/FAM214A acts in a conserved manner to regulate purine metabolism, promotes disease driven by hyperuricemia and associated tissue inflammation, and provides a potential novel target for uric acid-driven pathologies.


Asunto(s)
Proteínas de Drosophila , Gota , Hiperuricemia , Animales , Humanos , Ratones , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Estudio de Asociación del Genoma Completo , Gota/genética , Gota/complicaciones , Gota/metabolismo , Hiperuricemia/genética , Hiperuricemia/complicaciones , Hiperuricemia/metabolismo , Inflamación/genética , Inflamación/complicaciones , Purinas/metabolismo , Ácido Úrico/orina , Proteínas de Drosophila/genética
16.
Arthritis Rheum ; 62(7): 2004-12, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506365

RESUMEN

OBJECTIVE: Toll-like receptor 2 (TLR-2)/TLR-4-mediated innate immunity serves as a frontline antimicrobial host defense, but also modulates tissue remodeling and repair responses to endogenous ligands released during low-grade inflammation. We undertook the present study to assess whether the endogenous TLR-2/TLR-4 ligands low molecular weight hyaluronan (LMW-HA) and high mobility group box chromosomal protein 1 (HMGB-1), which are increased in osteoarthritic (OA) joints, drive procatabolic chondrocyte responses dependent on TLR-2 and TLR-4 signaling through the cytosolic adaptor myeloid differentiation factor 88 (MyD88). METHODS: We studied mature femoral head cap cartilage explants and immature primary knee articular chondrocytes from TLR-2/TLR-4-double-knockout, MyD88-knockout, and congenic wild-type mice. Generation of nitric oxide (NO), degradation of hyaluronan, release of HMGB-1, matrix metalloproteinase 3 (MMP-3), and MMP-13, and protein expression of type X collagen were assessed by Griess reaction and Western blotting analyses. Expression of messenger RNA for type II and type X collagen, MMP-13, and RUNX-2 was examined by real-time quantitative reverse transcription-polymerase chain reaction. RESULTS: Interleukin-1beta and TLR-2 and TLR-4 ligands induced both HMGB-1 release from chondrocytes and extracellular LMW-HA generation in normal chondrocytes. TLR-2/TLR-4(-/-) and MyD88(-/-) mouse cartilage explants and chondrocytes lost the capacity to mount procatabolic responses to both LMW-HA and HMGB-1, demonstrated by >95% suppression of NO production (P < 0.01), and attenuated induction of MMP-3 and MMP-13. Combined deficiency of TLR-2/TLR-4, or of MyD88 alone, also attenuated release of NO and blunted induction of MMP-3 and MMP-13 release. MyD88 was necessary for HMGB-1 and hyaluronidase 2 (which generates LMW-HA) to induce chondrocyte hypertrophy, which is implicated in OA progression. CONCLUSION: MyD88-dependent TLR-2/TLR-4 signaling is essential for procatabolic responses to LMW-HA and HMGB-1, and MyD88 drives chondrocyte hypertrophy. Therefore, LMW-HA and HMGB-1 act as innate immune cytokine-like signals with the potential to modulate chondrocyte differentiation and function in OA progression.


Asunto(s)
Condrocitos/metabolismo , Proteína HMGB2/metabolismo , Ácido Hialurónico/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Biomarcadores/metabolismo , Cartílago Articular/citología , Cartílago Articular/metabolismo , Aumento de la Célula , Condrocitos/citología , Femenino , Expresión Génica , Regulación de la Expresión Génica , Masculino , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peso Molecular , Factor 88 de Diferenciación Mieloide , Óxido Nítrico/metabolismo , Transducción de Señal
17.
Arthritis Rheum ; 62(7): 2170-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506351

RESUMEN

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.


Asunto(s)
Artritis Gotosa/metabolismo , Proteínas Portadoras/metabolismo , Proteínas/metabolismo , Secuencias Repetitivas de Aminoácido , Animales , Artritis Gotosa/inmunología , Células de la Médula Ósea , Proteínas Portadoras/inmunología , Células Cultivadas , Citocinas/metabolismo , Femenino , Proteínas Repetidas Ricas en Leucina , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Fagocitosis/efectos de los fármacos , Proteínas/genética , Proteínas/inmunología , Piel/efectos de los fármacos , Piel/patología , Ácido Úrico/inmunología , Ácido Úrico/toxicidad
18.
Methods Mol Biol ; 2245: 195-213, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33315204

RESUMEN

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.


Asunto(s)
Condrocitos/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/aislamiento & purificación , Mitocondrias/genética , Mitocondrias/metabolismo , Biogénesis de Organelos , Adenosina Trifosfato , Animales , Biomarcadores , Cartílago Articular/metabolismo , Regulación de la Expresión Génica , Humanos , Articulación de la Rodilla , Mutación , Osteoartritis/genética , Osteoartritis/metabolismo , Osteoartritis/patología , Procesamiento Proteico-Postraduccional , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Bioeng Transl Med ; 6(1): e10187, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33532587

RESUMEN

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.

20.
Immunol Cell Biol ; 88(1): 20-3, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19935768

RESUMEN

Gout is an inflammatory disease caused by the deposition of monosodium urate (MSU) crystals in the joint. Recent studies have significantly advanced our knowledge on the understanding of mechanisms underlying MSU crystal-induced inflammation. MSU crystals act as a 'danger signal' that can be recognized by pattern recognition receptors both at cell surface and cytoplasm, indicating the importance of innate immunity in gout. This review focuses on the critical role of intracellular NALP3 inflammasome in MSU crystal-induced inflammation.


Asunto(s)
Artritis Gotosa/inmunología , Proteínas Portadoras/inmunología , Inmunidad Innata , Animales , Humanos , Inflamación/inmunología , Espacio Intracelular/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Ácido Úrico/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA