Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 23(8)2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35457064

RESUMO

Camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome leads to diarthrodial joint arthropathy and is caused by the absence of lubricin (proteoglycan 4-PRG4), a surface-active mucinous glycoprotein responsible for lubricating articular cartilage. In this study, mice lacking the orthologous gene Prg4 served as a model that recapitulates the destructive arthrosis that involves biofouling of cartilage by serum proteins in lieu of Prg4. This study hypothesized that Prg4-deficient mice would demonstrate a quadruped gait change and decreased markers of mitochondrial dyscrasia, following intra-articular injection of both hindlimbs with recombinant human PRG4 (rhPRG4). Prg4-/- (N = 44) mice of both sexes were injected with rhPRG4 and gait alterations were studied at post-injection day 3 and 6, before joints were harvested for immunohistochemistry for caspase-3 activation. Increased stance and propulsion was shown at 3 days post-injection in male mice. There were significantly fewer caspase-3-positive chondrocytes in tibiofemoral cartilage from rhPRG4-injected mice. The mitochondrial gene Mt-tn, and myosin heavy (Myh7) and light chains (Myl2 and Myl3), known to play a cytoskeletal stabilizing role, were significantly upregulated in both sexes (RNA-Seq) following IA rhPRG4. Chondrocyte mitochondrial dyscrasias attributable to the arthrosis in CACP may be mitigated by IA rhPRG4. In a supporting in vitro crystal microbalance experiment, molecular fouling by albumin did not block the surface activity of rhPRG4.


Assuntos
Cartilagem Articular , Artropatias , Osteoartrite , Animais , Artropatia Neurogênica , Cartilagem Articular/metabolismo , Caspase 3 , Coxa Vara , Feminino , Marcha , Deformidades Congênitas da Mão , Injeções Intra-Articulares , Masculino , Camundongos , Camundongos Knockout , Proteoglicanas/metabolismo , Sinovite
2.
J Pharmacol Exp Ther ; 376(2): 222-230, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33239408

RESUMO

Gout is a chronic inflammatory arthritis caused by monosodium urate monohydrate (MSU) crystal deposits in joints of lower limbs. Phagocytic uptake of MSU crystals by joint-resident macrophages and recruited circulating monocytes results in IL-1ß expression and production. Current acute gout treatments have serious toxicities and suffer suboptimal clinical outcomes. Protein phosphatase 2A (PP2A) plays an important role in regulating signaling pathways relevant to inflammation. We hypothesized that innate immune danger signals, e.g., lipopolysaccharide (LPS) and soluble uric acid (sUA), prime human monocytes toward MSU crystal phagocytosis and that increased IL-1ß production mediated by a reduction in PP2A activity and restoring PP2A activity exerts an anti-inflammatory effect in this setting. Priming monocytes with LPS + sUA increased cytosolic pro-IL-1ß and mature IL-1ß and enhanced MSU crystal phagocytosis and its downstream IL-1ß expression (P < 0.001). A combination of LPS + sUA priming and MSU crystals reduced PP2A activity in monocytes by 60% (P = 0.013). PP2A catalytic subunit gene knockdown reduced PP2A activity and exacerbated MSU crystal-induced IL-1ß expression and secretion (P < 0.0001). Fingolimod (FTY720) and its active metabolite, fingolimod phosphate (FTY720-P), were evaluated for their ability to activate PP2A in human monocytes over 24 hours. FTY720 and FTY720-P activated PP2A to a similar extent, and maximal enzyme activity occurred at 24 hours for FTY720 and at 6 hours for FTY720-P. FTY720-P (2.5 µM) reduced pro-IL-1ß production and IL-1ß secretion in primed and MSU crystal-stimulated monocytes (P < 0.0001) without changing the magnitude of crystal phagocytosis. We conclude that PP2A is a promising new target in acute gout. SIGNIFICANCE STATEMENT: The activity of protein phosphatase 2A (PP2A) is implicated in the enhanced expression and production of IL-1ß by human monocytes in response to priming with soluble uric acid and lipopolysaccharide and phagocytosis of monosodium urate monohydrate (MSU) crystals. Fingolimod phosphate activates PP2A in human monocytes and reduces cytosolic pro-IL-1ß content and its conversion to biologically active IL-1ß in human monocytes exposed to MSU crystals.


Assuntos
Anti-Inflamatórios/farmacologia , Supressores da Gota/farmacologia , Interleucina-1beta/metabolismo , Monócitos/efeitos dos fármacos , Organofosfatos/farmacologia , Proteína Fosfatase 2/metabolismo , Esfingosina/análogos & derivados , Ácido Úrico/farmacologia , Humanos , Imunidade Inata/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/imunologia , Fagocitose , Proteína Fosfatase 2/efeitos dos fármacos , Esfingosina/farmacologia , Células THP-1
3.
J Immunol ; 200(2): 758-767, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29196459

RESUMO

Osteoarthritis (OA) is a low-grade chronic inflammatory joint disease. Innate immunity contributes to OA progression, mediated by TLR2 and TLR4. We evaluated the role of cluster determinant 44 (CD44), a transmembrane glycoprotein, in regulating TLR2-linked macrophage activation and resultant proinflammatory responses. TLR2 stimulation was performed on differentiated THP-1 macrophages in the presence or absence of a CD44-specific Ab or hyaluronan (HA). NF-κB nuclear translocation, IL-1 ß and TNF-α gene expression, and protein concentrations were determined. Anti-CD44 Ab and HA treatments reduced NF-κB translocation, IL-1ß and TNF-α expression, and production (p < 0.001). Inhibition of proinflammatory response in macrophages by HA was mediated by CD44. Protein phosphatase 2A mediated the reduction in NF-κB translocation by HA. CD44 knockdown reduced NF-κB nuclear translocation and downstream IL-1ß and TNF-α protein production following TLR2 receptor stimulation (p < 0.001). CD44+/+ murine bone marrow-derived macrophages produced higher TNF-α compared with CD44-/- macrophages following TLR2 stimulation (p < 0.01). HA dose-dependently inhibited TLR2-induced TNF-α production by murine bone marrow-derived macrophages (p < 0.001). OA synovial fluids (SF) stimulated TLR2 and TLR4 receptors and induced NF-κB translocation in THP-1 macrophages. Anti-CD44 Ab treatment significantly reduced macrophage activation by OA SF (p < 0.01). CD44 regulated TLR2 responses in human macrophages, whereby a reduction in CD44 levels or engagement of CD44 by its ligand (HA) or a CD44-specific Ab reduced NF-κB translocation and downstream proinflammatory cytokine production. A CD44-specific Ab reduced macrophage activation by OA SF, and CD44 is a potentially novel target in OA treatment.


Assuntos
Citocinas/genética , Citocinas/metabolismo , Receptores de Hialuronatos/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Receptor 2 Toll-Like/agonistas , Animais , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Receptores de Hialuronatos/genética , Ácido Hialurônico/metabolismo , Ligantes , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/metabolismo , Transporte Proteico , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
4.
Molecules ; 25(1)2019 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-31877739

RESUMO

Gout is an inflammatory arthritis due to the joint deposition of monosodium urate (MSU) crystals. Phagocytosis of MSU crystals by tissue macrophages results in the generation of reactive oxygen species (ROS) and production of inflammatory cytokines and chemokines. Colchicine use in gout is limited by severe toxicity. CD44 is a transmembrane glycoprotein that is highly expressed in tissue macrophages and may be involved in gout pathogenesis. The P6 peptide is a 20-amino acid residue peptide that binds to CD44. We hypothesized that the conjugation of colchicine to the P6 peptide would reduce its off-target cytotoxicity while preserving its anti-inflammatory effect. A modified version of P6 peptide and colchicine-P6 peptide conjugate were synthesized using Fmoc/tBu solid-phase and solution-phase chemistry, respectively. A glutaryl amide was used as a linker. The P6 peptide was evaluated for its binding to CD44, association, and internalization by macrophages. Cytotoxic effects of P6 peptide, colchicine, and colchicine-P6 peptide on macrophages were compared and the inhibition of ROS generation and interleukin-8 (IL-8) secretion in MSU-stimulated macrophages treated with P6 peptide, colchicine, or colchicine-P6 peptide was studied. We confirmed that the P6 peptide binds to CD44 and its association and internalization by macrophages were CD44-dependent. Colchicine (1, 10, and 25 µM) demonstrated a significant cytotoxic effect on macrophages while the P6 peptide and colchicine-P6 peptide conjugate (1, 10 and 25 µM) did not alter the viability of the macrophages. The P6 peptide (10 and 25 µM) reduced ROS generation and IL-8 secretion mediated by a reduction in MSU phagocytosis by macrophages. The colchicine-P6 peptide significantly reduced ROS generation and IL-8 secretion compared to the P6 peptide alone at 1 and 10 µM concentrations. Conjugation of colchicine to the P6 peptide reduced the cytotoxic effect of colchicine while preserving its anti-inflammatory activity.


Assuntos
Colchicina/farmacologia , Receptores de Hialuronatos/metabolismo , Imunoconjugados/farmacologia , Peptídeos/química , Colchicina/química , Humanos , Receptores de Hialuronatos/química , Imunoconjugados/química , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Modelos Biológicos , Estrutura Molecular , Fagocitose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Células THP-1 , Ácido Úrico/metabolismo
5.
Am J Physiol Cell Physiol ; 315(3): C432-C443, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29898378

RESUMO

Osteoarthritis (OA) is characterized by synovitis and synovial fibrosis. Synoviocytes are fibroblast-like resident cells of the synovium that are activated by transforming growth factor (TGF)-ß to proliferate, migrate, and produce extracellular matrix. Synoviocytes secrete hyaluronan (HA) and proteoglycan-4 (PRG4). HA reduces synovial fibrosis in vivo, and the Prg4-/- mouse exhibits synovial hyperplasia. We investigated the antifibrotic effects of increased intracellular cAMP in TGF-ß-stimulated human OA synoviocytes. TGF-ß1 stimulated collagen I (COL1A1), α-smooth muscle actin (α-SMA), tissue inhibitor of metalloproteinase (TIMP)-1, and procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) expression, and procollagen I, α-SMA, HA, and PRG4 production, migration, and proliferation of OA synoviocytes were measured. Treatment of OA synoviocytes with forskolin (10 µM) increased intracellular cAMP levels and reduced TGF-ß1-stimulated COL1A1, α-SMA, and TIMP-1 expression, with no change in PLOD2 expression. Forskolin also reduced TGF-ß1-stimulated procollagen I and α-SMA content as well as synoviocyte migration and proliferation. Forskolin (10 µM) increased HA secretion and PRG4 expression and production. A cell-permeant cAMP analog reduced COL1A1 and α-SMA expression and enhanced HA and PRG4 secretion by OA synoviocytes. HA and PRG4 reduced α-SMA expression and content, and PRG4 reduced COL1A1 expression and procollagen I content in OA synoviocytes. Prg4-/- synovium exhibited increased α-SMA, COL1A1, and TIMP-1 expression compared with Prg4+/+ synovium. Prg4-/- synoviocytes demonstrated strong α-SMA and collagen type I staining, whereas these were undetected in Prg4+/+ synoviocytes and were reduced with PRG4 treatment. We conclude that increasing intracellular cAMP levels in synoviocytes mitigates synovial fibrosis through enhanced production of HA and PRG4, possibly representing a novel approach for treatment of OA synovial fibrosis.


Assuntos
AMP Cíclico/metabolismo , Ácido Hialurônico/metabolismo , Osteoartrite/metabolismo , Proteoglicanas/metabolismo , Sinoviócitos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Idoso , Animais , Colforsina/farmacologia , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose/metabolismo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo , Sinoviócitos/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 110(15): 5852-7, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23530215

RESUMO

Osteoarthritis is a complex disease involving the mechanical breakdown of articular cartilage in the presence of altered joint mechanics and chondrocyte death, but the connection between these factors is not well established. Lubricin, a mucinous glycoprotein encoded by the PRG4 gene, provides boundary lubrication in articular joints. Joint friction is elevated and accompanied by accelerated cartilage damage in humans and mice that have genetic deficiency of lubricin. Here, we investigated the relationship between coefficient of friction and chondrocyte death using ex vivo and in vitro measurements of friction and apoptosis. We observed increases in whole-joint friction and cellular apoptosis in lubricin knockout mice compared with wild-type mice. When we used an in vitro bovine explant cartilage-on-cartilage bearing system, we observed a direct correlation between coefficient of friction and chondrocyte apoptosis in the superficial layers of cartilage. In the bovine explant system, the addition of lubricin as a test lubricant significantly lowered the static coefficient of friction and number of apoptotic chondrocytes. These results demonstrate a direct connection between lubricin, boundary lubrication, and cell survival and suggest that supplementation of synovial fluid with lubricin may be an effective treatment to prevent cartilage deterioration in patients with genetic or acquired deficiency of lubricin.


Assuntos
Apoptose , Condrócitos/patologia , Glicoproteínas/fisiologia , Proteoglicanas/fisiologia , Animais , Cartilagem/metabolismo , Caspase 3/metabolismo , Bovinos , Adesão Celular , Sobrevivência Celular , Genótipo , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Masculino , Camundongos , Camundongos Knockout , Osteoartrite/patologia , Osteoartrite/terapia , Proteoglicanas/genética , Resistência ao Cisalhamento , Estresse Mecânico
7.
Jt Comm J Qual Patient Saf ; 40(9): 398-407, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25252388

RESUMO

BACKGROUND: A study was conducted to evaluate the impact of a reengineered approach to electronic error reporting at a 719-bed multidisciplinary urban medical center. METHODS: The main outcome of interest was the monthly reported medication errors during the preimplementation (20 months) and postimplementation (26 months) phases. An interrupted time series analysis was used to describe baseline errors, immediate change following implementation of the current electronic error-reporting system (e-ERS), and trend of error reporting during postimplementation. Errors were categorized according to severity using the National Coordinating Council for Medication Error Reporting and Prevention (NCC MERP) Medication Error Index classifications. Reported errors were further analyzed by reporter and error site. RESULTS: During preimplementation, the monthly reported errors mean was 40.0 (95% confidence interval [CI]: 36.3-43.7). Immediately following e-ERS implementation, monthly reported errors significantly increased by 19.4 errors (95% CI: 8.4-30.5). The change in slope of reported errors trend was estimated at 0.76 (95% CI: 0.07-1.22). Near misses and no-patient-harm errors accounted for 90% of all errors, while errors that caused increased patient monitoring or temporary harm accounted for 9% and 1%, respectively. Nurses were the most frequent reporters, while physicians were more likely to report high-severity errors. Medical care units accounted for approximately half of all reported errors. CONCLUSION: Following the intervention, there was a significant increase in reporting of prevented errors and errors that reached the patient with no resultant harm. This improvement in reporting was sustained for 26 months and has contributed to designing and implementing quality improvement initiatives to enhance the safety of the medication use process.


Assuntos
Sistemas de Notificação de Reações Adversas a Medicamentos/organização & administração , Sistemas de Notificação de Reações Adversas a Medicamentos/estatística & dados numéricos , Hospitais Urbanos/organização & administração , Sistemas de Informação/organização & administração , Segurança do Paciente/estatística & dados numéricos , Hospitais com mais de 500 Leitos , Departamentos Hospitalares/estatística & dados numéricos , Hospitais Urbanos/estatística & dados numéricos , Humanos , Sistemas de Informação/estatística & dados numéricos , Internet , Erros de Medicação/prevenção & controle , Erros de Medicação/estatística & dados numéricos , Recursos Humanos em Hospital/estatística & dados numéricos , Melhoria de Qualidade/organização & administração , Melhoria de Qualidade/estatística & dados numéricos , Gestão de Riscos/organização & administração , Gestão de Riscos/estatística & dados numéricos
8.
Arthritis Rheum ; 64(4): 1162-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22127873

RESUMO

OBJECTIVE: To investigate whether cartilage degeneration is prevented or minimized in a rat model of anterior cruciate ligament (ACL) injury following a single dose-escalated intraarticular injection of lubricin derived from human synoviocytes in culture. METHODS: Unilateral ACL transection (ACLT) of the right hind limb was performed in Lewis rats (n = 56). Control animals underwent a capsulotomy alone, leaving the ACL intact (n = 11). Intraarticular injections (50 µl/injection) of phosphate buffered saline (PBS; n = 14 rats) and human synoviocyte lubricin (1,600 µg/ml; n = 14 rats) were performed on day 7 postsurgery. Animals were killed on day 70 postsurgery. Histologic specimens were immunoprobed for lubricin and sulfated glycosaminoglycans. Urinary C-telopeptide of type II collagen (CTX-II) levels were measured on days 35 and 70 postsurgery. Hind limb maximum applied force was determined using a variable resistor walkway to monitor quadruped gait asymmetries. RESULTS: Increased immunostaining for lubricin in the superficial zone and on the surface of cartilage was observed in lubricin-treated and control animals but not in PBS-treated or untreated animals with ACLT. On days 35 and 70 after surgery, urinary CTX-II levels in human synoviocyte lubricin-treated animals were lower than in untreated and PBS-treated animals (P < 0.005 and P < 0.001, respectively). Animals with ACLT treated with human synoviocyte lubricin and control animals distributed their weight equally between hind limbs compared to PBS-treated or untreated animals (P < 0.01). CONCLUSION: Our findings indicate that a single intraarticular injection of concentrated lubricin following ACLT reduces type II collagen degradation and improves weight bearing in the affected rat joint. These findings support the practice of tribosupplementation with lubricin for retarding cartilage degeneration and possibly the development of posttraumatic osteoarthritis.


Assuntos
Lesões do Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/efeitos dos fármacos , Cartilagem Articular/efeitos dos fármacos , Glicoproteínas/uso terapêutico , Traumatismos do Joelho/tratamento farmacológico , Lubrificação , Animais , Ligamento Cruzado Anterior/diagnóstico por imagem , Ligamento Cruzado Anterior/patologia , Cartilagem Articular/diagnóstico por imagem , Cartilagem Articular/patologia , Marcha , Glicoproteínas/administração & dosagem , Traumatismos do Joelho/diagnóstico por imagem , Traumatismos do Joelho/patologia , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Masculino , Radiografia , Ratos , Ratos Endogâmicos Lew
9.
Front Pharmacol ; 13: 1033520, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36467056

RESUMO

Background: Gout is a common arthritis, due to deposition of monosodium urate (MSU) crystals which results in IL-1ß secretion by tissue-resident macrophages. Xanthine oxidase (XO) catalyzes uric acid (UA) production and in the process, reactive oxygen species (ROS) are generated which contributes to NLRP3 inflammasome activation. Protein phosphatase 2A (PP2A) may be involved in regulating inflammatory pathways in macrophages. The objective of this study was to investigate whether PP2A regulates gout inflammation, mediated by XO activity modulation. We studied UA and ROS generations in MSU stimulated murine bone marrow derived macrophages (BMDMs) in response to fingolimod phosphate, a PP2A activator, and compared its anti-inflammatory efficacy to that of an XO inhibitor, febuxostat. Methods: BMDMs were stimulated with MSU, GM-CSF/IL-1ß or nigericin ± fingolimod (2.5 µM) or febuxostat (200 µM) and UA levels, ROS, XO, and PP2A activities, Xdh (XO) expression and secreted IL-1ß levels were determined. PP2A activity and IL-1ß in MSU stimulated BMDMs ± N-acetylcysteine (NAC) (10 µM) ± okadaic acid (a PP2A inhibitor) were also determined. M1 polarization of BMDMs in response to MSU ± fingolimod treatment was assessed by a combination of iNOS expression and multiplex cytokine assay. The in vivo efficacy of fingolimod was assessed in a murine peritoneal model of acute gout where peritoneal lavages were studied for pro-inflammatory classical monocytes (CMs), anti-inflammatory nonclassical monocytes (NCMs) and neutrophils by flow cytometry and IL-1ß by ELISA. Results: Fingolimod reduced intracellular and secreted UA levels (p < 0.05), Xdh expression (p < 0.001), XO activity (p < 0.001), ROS generation (p < 0.0001) and IL-1ß secretion (p < 0.0001), whereas febuxostat enhanced PP2A activity (p < 0.05). NAC treatment enhanced PP2A activity and reduced XO activity and PP2A restoration mediated NAC's efficacy as co-treatment with okadaic acid increased IL-1ß secretion (p < 0.05). Nigericin activated caspase-1 and reduced PP2A activity (p < 0.001) and fingolimod reduced caspase-1 activity in BMDMs (p < 0.001). Fingolimod reduced iNOS expression (p < 0.0001) and secretion of IL-6 and TNF-α (p < 0.05). Fingolimod reduced CMs (p < 0.0001), neutrophil (p < 0.001) and IL-1ß (p < 0.05) lavage levels while increasing NCMs (p < 0.001). Conclusion: Macrophage PP2A is inactivated in acute gout by ROS and a PP2A activator exhibited a broad anti-inflammatory effect in acute gout in vitro and in vivo.

10.
Healthcare (Basel) ; 10(9)2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-36141276

RESUMO

The study aims to assess office-based visit trends for lupus patients and evaluate their medication burden, chronic conditions, and comorbidities. This cross-sectional study used data from the National Ambulatory Medical Care Survey (NAMCS), a survey sample weighted to represent national estimates of outpatient visits. Adult patients diagnosed with lupus were included. Medications and comorbidities that were frequently recorded were identified and categorized. Descriptive statistics and bivariate analyses were used to characterize visits by sex, age, race/ethnicity, insurance type, region, and reason for visit. Comorbidities were identified using diagnosis codes documented at each encounter. There were 27,029,228 visits for lupus patients from 2006 to 2016, and 87% them were on or were prescribed medications. Most visits were for female (88%), white (79%), non-Hispanic (88%) patients with private insurance (53%). The majority of patients were seen for a chronic routine problem (75%), and 29% had lupus as the primary diagnosis. Frequent medications prescribed were hydroxychloroquine (30%), prednisone (23%), multivitamins (14%), and furosemide (9%). Common comorbidities observed included arthritis (88%), hypertension (25%), and depression (13%). Prescription patterns are reflective of comorbidities associated with lupus. By assessing medications most frequently prescribed and comorbid conditions among lupus patients, we showcase the complexity of disease management and the need for strategies to improve care.

11.
Arthritis Rheum ; 62(8): 2382-91, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20506144

RESUMO

OBJECTIVE: To investigate whether cartilage degeneration is prevented or minimized following intraarticular injections of lubricin derived from human synoviocytes in culture, recombinant human PRG4 (rhPRG4), or human synovial fluid (SF) in a rat model of anterior cruciate ligament (ACL) injury. METHODS: Unilateral ACL transection (ACLT) was performed in Lewis rats (n = 45). Nine animals were left untreated. The remaining rats were given intraarticular injections (50 microl/injection) of either phosphate buffered saline (PBS) (n = 9), human synoviocyte lubricin (200 microg/ml; n = 9), rhPRG4 (200 microg/ml; n = 9), or human SF lubricin (200 microg/ml; n = 9) twice weekly beginning on day 7 after injury. Joints were harvested on day 32 after injury. Histologic analysis was performed using Safranin O-fast green staining, and articular cartilage degeneration was graded using the Osteoarthritis Research Society International (OARSI)-modified Mankin criteria. Histologic specimens were immunoprobed for lubricin and sulfated glycosaminoglycans. A 24-hour urine collection was performed on days 17 and 29 postinjury, and urinary C-terminal telopeptide of type II collagen (CTX-II) levels were measured. RESULTS: Treatment with human synoviocyte lubricin resulted in significantly lower OARSI scores for cartilage degeneration compared with no treatment or PBS treatment (P < 0.05). Increased immunostaining for lubricin in the superficial zone chondrocytes and on the surface of cartilage was observed in lubricin-treated, but not untreated or PBS-treated, joints. On day 17, urinary CTX-II levels in human synoviocyte lubricin- and human SF lubricin-treated animals were significantly lower than those in untreated animals (P = 0.005 and P = 0.002, respectively) and in PBS-treated animals (P = 0.002 and P < 0.001, respectively). CONCLUSION: After treatment with any of the 3 types of lubricin evaluated in this study, a reduction in cartilage damage following ACLT was evident, combined with a reduction in type II collagen degradation. Our findings indicate that intraarticular lubricin injection following an ACL injury may be beneficial in retarding the degeneration of cartilage and the development of posttraumatic OA.


Assuntos
Ligamento Cruzado Anterior/patologia , Cartilagem Articular/patologia , Condrócitos/patologia , Citoproteção/efeitos dos fármacos , Glicoproteínas/farmacologia , Animais , Ligamento Cruzado Anterior/efeitos dos fármacos , Ligamento Cruzado Anterior/metabolismo , Lesões do Ligamento Cruzado Anterior , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Colágeno Tipo II/metabolismo , Glicoproteínas/metabolismo , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Injeções Intra-Articulares , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/metabolismo , Articulação do Joelho/patologia , Masculino , Distribuição Aleatória , Ratos , Ratos Endogâmicos Lew
12.
Int J Qual Health Care ; 23(1): 36-43, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21084323

RESUMO

OBJECTIVE: To reduce the probability of failure in the oral chemotherapy order, review and administration process and to reduce oral chemotherapy-related prescribing errors intercepted by clinical pharmacists prior to reaching the patient. DESIGN: A before-and-after cohort study. SETTING: A 719-bed multidisciplinary tertiary care institution with a pediatric division and an outpatient cancer center. PARTICIPANTS: A multidisciplinary team characterized key elements of the oral chemotherapy process using healthcare failure modes and effects analysis (HFMEA). INTERVENTION(S): Oral chemotherapy computerized provider order entry (CPOE) was developed and implemented. MAIN OUTCOME MEASURE(S): Pharmacist-intercepted oral chemotherapy prescribing errors over a 24-month period (before) and over a 6-month period (after) were analyzed according to the error type (errors in clinical decision making, errors in transcription or errors related to prescribing policy). The incidence of prescribing errors prior to and following CPOE implementation was compared by calculating the odds ratio (OR) and the 95% confidence interval (CI). RESULTS: HFMEA hazard analysis revealed seven potential failure modes, with the highest hazard scores in the prescribing and administration components of the process. CPOE implementation significantly (P= 0.023) reduced prescribing error risk by 69% [OR (95% CI) = 0.31 (0.11-0.86)] and eliminated certain types of errors that can lead to significant patient harm. CONCLUSIONS: Prescribing oral chemotherapy is a failure mode with significant risk of inducing patient harm. CPOE is effective in reducing prescribing errors of oral chemotherapy and should be considered part of a fail-safe process to improve safety.


Assuntos
Antineoplásicos/administração & dosagem , Sistemas de Registro de Ordens Médicas/organização & administração , Erros de Medicação/prevenção & controle , Serviço de Farmácia Hospitalar/organização & administração , Gestão da Segurança/organização & administração , Administração Oral , Estudos de Coortes , Hospitais com mais de 500 Leitos/estatística & dados numéricos , Humanos , Sistemas de Registro de Ordens Médicas/estatística & dados numéricos , Erros de Medicação/classificação , Erros de Medicação/estatística & dados numéricos , Serviço de Farmácia Hospitalar/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Gestão da Segurança/estatística & dados numéricos
13.
Front Immunol ; 12: 771677, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34992596

RESUMO

Objectives: To compare phagocytic activities of monocytes in peripheral blood mononuclear cells (PBMCs) from acute gout patients and normal subjects, examine monosodium urate monohydrate (MSU) crystal-induced IL-1ß secretion ± recombinant human proteoglycan 4 (rhPRG4) or interleukin-1 receptor antagonist (IL-1RA), and study the anti-inflammatory mechanism of rhPRG4 in MSU stimulated monocytes. Methods: Acute gout PBMCs were collected from patients in the Emergency Department and normal PBMCs were obtained from a commercial source. Monocytes in PBMCs were identified by flow cytometry. PBMCs were primed with Pam3CSK4 (1µg/mL) for 24h and phagocytic activation of monocytes was determined using fluorescently labeled latex beads. MSU (200µg/mL) stimulated IL-1ß secretion was determined by ELISA. Reactive oxygen species (ROS) generation in monocytes was determined fluorometrically. PBMCs were incubated with IL-1RA (250ng/mL) or rhPRG4 (200µg/mL) and bead phagocytosis by monocytes was determined. THP-1 monocytes were treated with MSU crystals ± rhPRG4 and cellular levels of NLRP3 protein, pro-IL-1ß, secreted IL-1ß, and activities of caspase-1 and protein phosphatase-2A (PP2A) were quantified. The peritoneal influx of inflammatory and anti-inflammatory monocytes and neutrophils in Prg4 deficient mice was studied and the impact of rhPRG4 on immune cell trafficking was assessed. Results: Enhanced phagocytic activation of gout monocytes under basal conditions (p<0.001) was associated with ROS generation and MSU stimulated IL-1ß secretion (p<0.05). rhPRG4 reduced bead phagocytosis by normal and gout monocytes compared to IL-1RA and both treatments were efficacious in reducing IL-1ß secretion (p<0.05). rhPRG4 reduced pro-IL-1ß content, caspase-1 activity, conversion of pro-IL-1ß to mature IL-1ß and restored PP2A activity in monocytes (p<0.05). PP2A inhibition reversed rhPRG4's effects on pro-IL-1ß and mature IL-1ß in MSU stimulated monocytes. Neutrophils accumulated in peritoneal cavities of Prg4 deficient mice (p<0.01) and rhPRG4 treatment reduced neutrophil accumulation and enhanced anti-inflammatory monocyte influx (p<0.05). Conclusions: MSU phagocytosis was higher in gout monocytes resulting in higher ROS and IL-1ß secretion. rhPRG4 reduced monocyte phagocytic activation to a greater extent than IL-1RA and reduced IL-1ß secretion. The anti-inflammatory activity of rhPRG4 in monocytes is partially mediated by PP2A, and in vivo, PRG4 plays a role in regulating the trafficking of immune cells into the site of a gout flare.


Assuntos
Anti-Inflamatórios/uso terapêutico , Gota/tratamento farmacológico , Interleucina-1beta/imunologia , Proteoglicanas/uso terapêutico , Acetilcisteína/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anti-Inflamatórios/farmacologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Gota/imunologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/antagonistas & inibidores , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fagocitose/efeitos dos fármacos , Proteína Fosfatase 2/imunologia , Proteoglicanas/genética , Proteoglicanas/farmacologia , Espécies Reativas de Oxigênio/imunologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Células THP-1 , Ácido Úrico
14.
Arthritis Res Ther ; 23(1): 241, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34521469

RESUMO

BACKGROUND: Synovial macrophages perform a multitude of functions that include clearance of cell debris and foreign bodies, tissue immune surveillance, and resolution of inflammation. The functional diversity of macrophages is enabled by distinct subpopulations that express unique surface markers. Proteoglycan-4 (PRG4) is an important regulator of synovial hyperplasia and fibrotic remodeling, and the involvement of macrophages in PRG4's synovial role is yet to be defined. Our objectives were to study the PRG4's importance to macrophage homeostatic regulation in the synovium and infiltration of pro-inflammatory macrophages in acute synovitis and investigate whether macrophages mediated synovial fibrosis in Prg4 gene-trap (Prg4GT/GT) murine knee joints. METHODS: Macrophage phenotyping in Prg4GT/GT and Prg4+/+ joints was performed by flow cytometry using pan-macrophage markers, e.g., CD11b, F4/80, and surface markers of M1 macrophages (CD86) and M2 macrophages (CD206). Characterizations of the various macrophage subpopulations were performed in 2- and 6-month-old animals. The expression of inflammatory markers, IL-6, and iNOS in macrophages that are CD86+ and/or CD206+ was studied. The impact of Prg4 recombination on synovial macrophage populations of 2- and 6-month-old animals and infiltration of pro-inflammatory macrophages in response to a TLR2 agonist challenge was determined. Macrophages were depleted using liposomal clodronate and synovial membrane thickness, and the expression of fibrotic markers α-SMA, PLOD2, and collagen type I (COL-I) was assessed using immunohistochemistry. RESULTS: Total macrophages in Prg4GT/GT joints were higher than Prg4+/+ joints (p<0.0001) at 2 and 6 months, and the percentages of CD86+/CD206- and CD86+/CD206+ macrophages increased in Prg4GT/GT joints at 6 months (p<0.0001), whereas the percentage of CD86-/CD206+ macrophages decreased (p<0.001). CD86+/CD206- and CD86+/CD206+ macrophages expressed iNOS and IL-6 compared to CD86-/CD206+ macrophages (p<0.0001). Prg4 re-expression limited the accumulation of CD86+ macrophages (p<0.05) and increased CD86-/CD206+ macrophages (p<0.001) at 6 months. Prg4 recombination attenuated synovial recruitment of pro-inflammatory macrophages in 2-month-old animals (p<0.001). Clodronate-mediated macrophage depletion reduced synovial hyperplasia, α-SMA, PLOD2, and COL-I expressions in the synovium (p<0.0001). CONCLUSIONS: PRG4 regulates the accumulation and homeostatic balance of macrophages in the synovium. In its absence, the synovium becomes populated with M1 macrophages. Furthermore, macrophages exert an effector role in synovial fibrosis in Prg4GT/GT animals.


Assuntos
Ativação de Macrófagos , Macrófagos , Proteoglicanas , Animais , Inflamação , Camundongos , Proteoglicanas/genética , Membrana Sinovial
15.
Sci Rep ; 10(1): 5748, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238827

RESUMO

Gout is a chronic arthritis caused by the deposition of poorly soluble monosodium urate monohydrate (MSU) crystals in peripheral joints. Resident macrophages initiate inflammation in response to MSU mediated by NF-κB nuclear translocation and NLRP3 inflammasome activation. We investigated the role of CD44, a transmembrane receptor, in mediating MSU phagocytosis by macrophages. We used an antibody that sheds the extracellular domain (ECD) of CD44 to study the role of the receptor and its associated protein phosphatase 2A (PP2A) in macrophage activation. We also studied the significance of CD44 in mediating MSU inflammation in-vivo. Cd44-/- BMDMs showed reduced MSU phagocytosis, LDH release, IL-1ß expression and production compared to Cd44+/+ BMDMs. Elevated CD44 staining was detected intracellularly and CD44 colocalized with α-tubulin as a result of MSU exposure and ECD-shedding reduced MSU phagocytosis in murine and human macrophages. Anti-CD44 antibody treatment reduced NF-κB p65 subunit nuclear levels, IL-1ß expression, pro-IL-1ß and IL-8 production in MSU stimulated THP-1 macrophages (p < 0.01). The effect of the antibody was mediated by an enhancement in PP2A activity. CD44 ECD-shedding reduced the conversion of procaspase-1 to active caspase-1, caspase-1 activity and resultant generation of mature IL-1ß in macrophages. Neutrophil and monocyte influx and upregulated production of IL-1ß was evident in wildtype mice. MSU failed to trigger neutrophil and monocyte recruitment in Cd44-/- mice and lower IL-1ß levels were detected in peritoneal lavages from Cd44-/- mice (p < 0.01). Anti-CD44 antibody treatment reduced neutrophil and monocyte recruitment and resulted in reduced lavage IL-1ß levels in the same model. CD44 plays a biologically significant role in mediating phagocytosis of MSU and downstream inflammation and is a novel target in gout treatment.


Assuntos
Artrite Gotosa/imunologia , Receptores de Hialuronatos/imunologia , Macrófagos/imunologia , Fagocitose , Ácido Úrico/imunologia , Animais , Artrite Gotosa/patologia , Linhagem Celular , Células Cultivadas , Cristalização , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos , Ácido Úrico/análise
16.
Crit Care Explor ; 2(6): e0126, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32695993

RESUMO

OBJECTIVES: Sepsis is a leading cause of death in the United States. Putative targets to prevent systemic inflammatory response syndrome include antagonism of toll-like receptors 2 and 4 and CD44 receptors in vascular endothelial cells. Proteoglycan-4 is a mucinous glycoprotein that interacts with CD44 and toll-like receptor 4 resulting in a blockade of the NOD-like receptor pyrin domain-containing-3 pathway. We hypothesized that endothelial cells induced into a sepsis phenotype would have less interleukin-6 expression after recombinant human proteoglycan 4 treatment in vitro. DESIGN: Enzyme-linked immunosorbent assay and reverse transcriptase-quantitative polymerase chain reaction to measure interleukin-6 protein and gene expression. SETTING: Research laboratory. SUBJECTS: Human umbilical vascular endothelial cells, human lung microvascular endothelial cells, and transgenic mouse (wild type) (Cd44 +/+/Prg4 +/+), Cd44 -/- (Cd44 tm1Hbg Prg4 +/+), Prg4 GT/GT (Cd44 +/+ Prg4 tm2Mawa/J), and double knockout (Cd44 tm1Hbg Prg4 tm2Mawa/J) lung microvascular endothelial cells. INTERVENTIONS: Cells were treated with 100 or 250 ng/mL lipopolysaccharide-Escherichia coli K12 and subsequently treated with recombinant human proteoglycan 4 after 30 minutes. Interleukin-6 levels in conditioned media were measured via enzyme-linked immunosorbent assay and gene expression was measured via reverse transcriptase-quantitative polymerase chain reaction with ΔΔ-Ct analysis. Additionally, human umbilical vascular endothelial cells and human lung microvascular endothelial cells were treated with 1:10 diluted plasma from 15 patients with sepsis in culture media. After 30 minutes, either 50 or 100 µg/mL recombinant human proteoglycan 4 was administered. Interleukin-6 protein and gene expression were assayed. Proteoglycan 4 levels were also compared between control and sepsis patient plasma. MEASUREMENTS AND MAIN RESULTS: Human umbilical vascular endothelial cell, human lung microvascular endothelial cell, and mouse lung microvascular endothelial cell treated with lipopolysaccharide had significantly increased interleukin-6 protein compared with controls. Recombinant human proteoglycan-4 significantly reduced interleukin-6 in human and mouse endothelial cells. Interleukin-6 gene expression was significantly increased after lipopolysaccharide treatment compared with controls. This response was reversed by 50 or 100 µg/mL recombinant human proteoglycan-4 in 80% of sepsis samples in human umbilical vascular endothelial cells and in 60-73% in human lung microvascular endothelial cells. In Cd44 -/- genotypes of the mouse lung microvascular endothelial cells, recombinant human proteoglycan-4 significantly reduced interleukin-6 protein levels after lipopolysaccharide treatment, indicating that Cd44 is not needed for recombinant human proteoglycan-4 to have an effect in a toll-like receptor 4 agonist inflammation model. Patient sepsis samples had higher plasma levels of native proteoglycan-4 than controls. INTERPRETATION AND CONCLUSIONS: Recombinant human proteoglycan-4 is a potential adjunct therapy for sepsis patients and warrants future in vivo model studies.

17.
Arthritis Res Ther ; 22(1): 113, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32404156

RESUMO

BACKGROUND: Synovial tissue fibrosis is common in advanced OA with features including the presence of stress fiber-positive myofibroblasts and deposition of cross-linked collagen type-I. Proteoglycan-4 (PRG4) is a mucinous glycoprotein secreted by synovial fibroblasts and is a major component of synovial fluid. PRG4 is a ligand of the CD44 receptor. Our objective was to examine the role of PRG4-CD44 interaction in regulating synovial tissue fibrosis in vitro and in vivo. METHODS: OA synoviocytes were treated with TGF-ß ± PRG4 for 24 h and α-SMA content was determined using immunofluorescence. Rhodamine-labeled rhPRG4 was incubated with OA synoviocytes ± anti-CD44 or isotype control antibodies and cellular uptake of rhPRG4 was determined following a 30-min incubation and α-SMA expression following a 24-h incubation. HEK-TGF-ß cells were treated with TGF-ß ± rhPRG4 and Smad3 phosphorylation was determined using immunofluorescence and TGF-ß/Smad pathway activation was determined colorimetrically. We probed for stress fibers and focal adhesions (FAs) in TGF-ß-treated murine fibroblasts and fibroblast migration was quantified ± rhPRG4. Synovial expression of fibrotic markers: α-SMA, collagen type-I, and PLOD2 in Prg4 gene-trap (Prg4GT) and recombined Prg4GTR animals were studied at 2 and 9 months of age. Synovial expression of α-SMA and PLOD2 was determined in 2-month-old Prg4GT/GT&Cd44-/- and Prg4GTR/GTR&Cd44-/- animals. RESULTS: PRG4 reduced α-SMA content in OA synoviocytes (p < 0.001). rhPRG4 was internalized by OA synoviocytes via CD44 and CD44 neutralization attenuated rhPRG4's antifibrotic effect (p < 0.05). rhPRG4 reduced pSmad3 signal in HEK-TGF-ß cells (p < 0.001) and TGF-ß/Smad pathway activation (p < 0.001). rhPRG4 reduced the number of stress fiber-positive myofibroblasts, FAs mean size, and cell migration in TGF-ß-treated NIH3T3 fibroblasts (p < 0.05). rhPRG4 inhibited fibroblast migration in a macrophage and fibroblast co-culture model without altering active or total TGF-ß levels. Synovial tissues of 9-month-old Prg4GT/GT animals had higher α-SMA, collagen type-I, and PLOD2 (p < 0.001) content and Prg4 re-expression reduced these markers (p < 0.01). Prg4 re-expression also reduced α-SMA and PLOD2 staining in CD44-deficient mice. CONCLUSION: PRG4 is an endogenous antifibrotic modulator in the joint and its effect on myofibroblast formation is partially mediated by CD44, but CD44 is not required to demonstrate an antifibrotic effect in vivo.


Assuntos
Diferenciação Celular , Miofibroblastos/citologia , Proteoglicanas/genética , Membrana Sinovial , Idoso , Animais , Feminino , Fibroblastos , Fibrose , Humanos , Receptores de Hialuronatos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Células NIH 3T3
18.
J Orthop Res ; 37(2): 386-396, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30488470

RESUMO

Treatment of the injured joint with rhPRG4 is based on recent observations that inflammation diminishes expression of native PRG4. Re-establishing lubrication between pressurized and sliding cartilage surfaces during locomotion promotes the nascent expression of PRG4 and thus intra-articular (IA) treatment strategies should be supported by pharmacokinetic evidence establishing the residence time of rhPRG4. A total of 21 Yucatan minipigs weighing ∼55 kg each received 4 mg of 131 I-rhPRG4 delivered by IA injection 5 days following surgical ACL transection. Animals were sequentially euthanized following IA rhPRG4 at 10 min (time zero), 24, 72 h, 6, 13 and 20 days later. The decay of the 131 I-rhPRG4 was measured relative to a non-injected aliquot and normalized to the weight of cartilage samples, menisci and synovium, and known cartilage volumes from each compartment surface obtained from representative Yucatan minipig knees. Decay of 131 I-rhPRG4 from joint tissues best fit a two-compartment model with an α half-life (t1/2α ) of 11.28 h and ß half-life (t1/2ß ) of 4.81 days. The tibial and femoral cartilage, meniscii, and synovium retained 7.7% of dose at 24 h. High concentrations of rhPRG4 were found in synovial fluid (SF) that was non-aspiratable and resided on the articular surfaces, removable by irrigation, at 10 min following 131 I-rhPRG4 injection. Synovial fluid K21 exceeded K12 and SF t1/2ß was 28 days indicating SF is the reservoir for rhPRG4 following IA injection. Clinical Significance: rhPRG4 following IA delivery in a traumatized joint populates articular surfaces for a considerable period and may promote the native expression of PRG4. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:386-396, 2019.


Assuntos
Lesões do Ligamento Cruzado Anterior/metabolismo , Glicoproteínas/farmacocinética , Articulação do Joelho/metabolismo , Proteoglicanas/farmacocinética , Animais , Feminino , Radioisótopos do Iodo , Masculino , Suínos , Porco Miniatura
19.
J Pharm Pract Res ; 48(1): 65-71, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36815097

RESUMO

Background: A multidisciplinary approach is recommended for the management of type 2 diabetes mellitus (DM). Aim: To evaluate the impact of a pharmacist intervention on haemoglobin A1c (HbA1c), systolic blood pressure (SBP), diastolic blood pressure (DBP), and diabetes-related hospitalisations in an underserved cohort with unmanaged type 2 DM. Methods: This analysis was a retrospective cohort study. Criteria for inclusion were persons with unmanaged type 2 DM defined as HbA1c values ≥8% at time of enrolment, ≥18 years old, and enrolment in a pharmacist-managed clinic for ≥12 months. Pre- and post-intervention differences in HbA1c, SBP and DBP values were assessed using repeated measures analysis of variance (ANOVA). The risk of diabetes-related hospitalisations was estimated during the 12 months prior and during the 12 months post-intervention, and the relative risk (RR) was calculated. Results: Mean HbA1c values at 3, 6 and 12 months post-intervention were lower than baseline values (p < 0.05). There was no significant difference in mean HbA1c values at 6 or 12 months compared to 3 months post intervention. Mean SBP values at 3, 6 and 12 months were lower than baseline (p < 0.05). Likewise, mean DBP values at 6 and 12 months were lower than baseline (p < 0.05). The estimated RR of diabetes-related hospitalisations was 0.40 (95% CI: 0.20-0.83; p = 0.013). Conclusion: Enrolment in a pharmacist-managed diabetes program was associated with a significant reduction in HbA1c, SBP and DBP and reduction in risk of diabetes-related hospitalisations in an underserved cohort of patients with diabetes over a 12-month period.

20.
Arthritis Res Ther ; 20(1): 192, 2018 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-30157934

RESUMO

BACKGROUND: Gout is an inflammatory arthritis caused by monosodium urate monohydrate (MSU) crystals' joint deposition. MSU phagocytosis by resident macrophages is a key step in gout pathogenesis. MSU phagocytosis triggers nuclear factor kappa B (NFκB) activation and production of cytokines and chemokines. Proteoglycan-4 (PRG4) is a glycoprotein produced by synovial fibroblasts and exerts an anti-inflammatory effect in the joint mediated by its interaction with cell surface receptor CD44. PRG4 also binds and antagonizes TLR2 and TLR4. The objective of this study is to evaluate the efficacy of recombinant human PRG4 (rhPRG4) in suppressing MSU-induced inflammation and mechanical allodynia in vitro and in vivo. METHODS: THP-1 macrophages were incubated with MSU crystals ± rhPRG4 or bovine submaxillary mucin (BSM), and crystal phagocytosis, cytokines and chemokines expression and production were determined. NFκB p65 subunit nuclear translocation, NLRP3 induction, caspase-1 activation and conversion of proIL-1ß to mature IL-1ß were studied. MSU phagocytosis by Prg4+/+ and Prg4-/- peritoneal macrophages was determined in the absence or presence of rhPRG4, BSM, anti-CD44, anti-TLR2, anti-TLR4 and isotype control antibodies. Rhodamine-labeled rhPRG4 was incubated with murine macrophages and receptor colocalization studies were performed. Lewis rats underwent intra-articular injection of MSU crystals followed by intra-articular treatment with PBS or rhPRG4. Weight bearing and SF myeloperoxidase activities were determined. RESULTS: rhPRG4 reduced MSU crystal phagocytosis at 4 h (p < 0.01) and IL-1ß, TNF-α, IL-8 and MCP-1 expression and production at 6 h (p < 0.05). BSM did not alter MSU phagocytosis or IL-1ß production in human and murine macrophages. rhPRG4 treatment reduced NFκB nuclear translocation, NLRP3 expression, caspase-1 activation and generation of mature IL-1ß (p < 0.05). MSU-stimulated IL-1ß production was higher in Prg4-/- macrophages compared to Prg4+/+ macrophages (p < 0.001). rhPRG4, anti-CD44, anti-TLR2 and anti-TLR4 antibody treatments reduced MSU phagocytosis and IL-1ß production in murine macrophages (p < 0.05). rhPRG4 preferentially colocalized with CD44 on Prg4-/- peritoneal macrophages compared to TLR2 or TLR4 (p < 0.01). rhPRG4 normalized weight bearing and reduced SF myeloperoxidase activity compared to PBS in vivo. CONCLUSION: rhPRG4 inhibits MSU crystal phagocytosis and exhibits an anti-inflammatory and anti-nociceptive activity in vitro and in vivo. rhPRG4's anti-inflammatory mechanism may be due to targeting CD44 on macrophages.


Assuntos
Citocinas/metabolismo , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , NF-kappa B/metabolismo , Fagocitose/efeitos dos fármacos , Proteoglicanas/farmacologia , Ácido Úrico/farmacologia , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Quimiocinas/genética , Quimiocinas/metabolismo , Cristalização , Citocinas/genética , Humanos , Receptores de Hialuronatos/antagonistas & inibidores , Receptores de Hialuronatos/imunologia , Receptores de Hialuronatos/metabolismo , Inflamassomos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Knockout , Proteoglicanas/genética , Proteoglicanas/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Células THP-1 , Ácido Úrico/química , Ácido Úrico/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA