Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Thromb Haemost ; 21(9): 2390-2404, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37116753

RESUMO

BACKGROUND: Mechanisms of iron clearance from hemophilic joints are unknown. OBJECTIVES: To better understand mechanisms of iron clearance following joint bleeding in a mouse model of hemophilia. METHODS: Hemarthrosis was induced by subpatellar puncture in factor VIII (FVIII)-deficient (FVII-/-) mice, +/- periprocedural recombinant human FVIII, and hypocoagulable (HypoBALB/c) mice. HypoBALB/c mice experienced transient FVIII deficiency (anti-FVIII antibody) at the time of injury combined with warfarin-induced hypocoagulability. Synovial tissue was harvested weekly up to 6 weeks after injury for histological analysis, ferric iron and macrophage accumulation (CD68), blood and lymphatic vessel remodeling (αSMA; LYVE1). Synovial RNA sequencing was performed for FVIII-/- mice at days 0, 3, and 14 after injury to quantify expression changes of iron regulators and lymphatic markers. RESULTS: Bleed volumes were similar in FVIII-/- and HypoBALB/c mice. However, pronounced and prolonged synovial iron accumulation colocalizing with macrophages and impaired lymphangiogenesis were detected only in FVIII-/- mice and were prevented by periprocedural FVIII. Gene expression changes involved in iron handling (some genes with dual roles in inflammation) and lymphatic markers supported proinflammatory milieu with iron retention and disturbed lymphangiogenesis. CONCLUSION: Accumulation and delayed clearance of iron-laden macrophages were associated with defective lymphangiogenesis after hemarthrosis in FVIII-/- mice. The absence of such findings in HypoBALB/c mice suggests that intact lymphatics are required for removal of iron-laden macrophages and that these processes depend on FVIII availability. Studies to elucidate the biological mechanisms of disturbed lymphangiogenesis in hemophilia appear critical to develop new therapeutic targets.


Assuntos
Hemofilia A , Hemostáticos , Camundongos , Humanos , Animais , Fator VIII/genética , Fator VIII/metabolismo , Hemartrose/metabolismo , Hemofilia A/terapia , Linfangiogênese , Modelos Animais de Doenças , Ferro
2.
Clin Orthop Relat Res ; 481(8): 1634-1647, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37036937

RESUMO

BACKGROUND: Hemophilic arthropathy can cause recurrent hemarthroses and severe damage to the synovium and articular cartilage. Previous studies have shown that vascular endothelial growth factor (VEGF) plays an essential role in neoangiogenesis. Bevacizumab, a monoclonal VEGF inhibitor, is used clinically to prevent angiogenesis. However, its effects on hemophilic arthropathy are unknown. QUESTIONS/PURPOSES: Using a hemophilic arthropathy rabbit model, we asked: Does an intra-articular injection of bevacizumab (1) inhibit VEGF, (2) decrease signal intensity in dynamic contrast-enhanced MRI (DCE-MRI) as an assessment of capillary permeability and neoangiogenesis, (3) reduce cartilage damage, (4) reduce synovial changes, and (5) affect macroscopic changes during the development of hemophilic arthropathy? METHODS: Twenty-five male New Zealand rabbits were divided into four groups. Eight knees from four rabbits were used as the control group. We used an established animal model for hemophilic arthropathy in the remaining 21 rabbits. Animals were assigned randomly to three groups with seven rabbits in each group. One group was used to establish mild arthropathy, and the other two were used to establish severe arthropathy. Autologous blood from the rabbits' ears was injected into the right and left knees twice per week for 8 weeks to represent mild arthropathy and for 16 weeks to represent severe arthropathy. In the mild arthropathy group, bevacizumab was injected into the right knee once every 2 weeks. Bevacizumab was injected into the right knee of rabbits in one of the severe arthropathy groups once every 2 weeks for 16 weeks, and intra-articular bevacizumab injections were administered to the right knees of rabbits in the other severe arthropathy group once every 2 weeks after the eighth week. An equal volume of 0.9% saline was injected into the left knee of rabbits in all arthropathy groups. To explore the efficacy of bevacizumab, joint diameters were quantitatively measured, and cartilage and synovial changes were examined. Degeneration of articular cartilage was evaluated with the semiquantitative Osteoarthritis Research Society International grading system. Synovial damage was analyzed with a semiquantitative microscopic scoring system. In addition, we evaluated perfusion and angiogenesis using DCE-MRI (quantitative signal intensity changes). Immunohistochemical testing was used to measure VEGF levels (analyzed by Western blotting). RESULTS: Intra-articular bevacizumab treatment inhibited VEGF in our rabbit model of hemophilic arthropathy. VEGF protein expression levels were lower in the mild arthropathy group that received intra-articular bevacizumab (0.89 ± 0.45) than the mild arthropathy control group (1.41 ± 0.61) (mean difference -0.52 [95% CI -0.898 to -0.143]; p = 0.02). VEGF levels were lower in the severe arthropathy group that received treatment for 16 weeks (0.94 ± 0.27) than in the control knees (1.49 ± 0.36) (mean difference -0.55 [95% CI -0.935 to -0.161]; p = 0.01). In the severe arthropathy group, the Osteoarthritis Research Society International score indicating cartilage damage was lower in the group that received intra-articular bevacizumab treatment from the beginning than in the control group (median 17 [range 13 to 18] versus 18 [range 17 to 20]; difference of medians 1; p = 0.02). Additionally, the scores indicated synovial damage was lower in the group that received intra-articular bevacizumab treatment from the beginning than the control group (median 5 [range 4 to 9] versus 9 [range 8 to 12]; difference of medians 4; p = 0.02). The mean of mean values for signal intensity changes was higher in the nontreated severe groups than in the group of healthy knees. The signal intensity changes were higher in the severe arthropathy control groups (Groups BC and CC) (median 311.6 [range 301.4 to 361.2] and 315.1 [range 269.7 to 460.4]) than in the mild arthropathy control group (Group AC) (median 234.1 [range 212.5 to 304.2]; difference of medians 77.5 and 81, respectively; p = 0.02 and p = 0.04, respectively). In the severe arthropathy group, discoloration caused by hemosiderin deposition in the cartilage and synovium was more pronounced than in the mild arthropathy group. In the severe arthropathy group treated with intra-articular bevacizumab, joint diameters were smaller than in the control group (Group BT median 12.7 mm [range 12.3 to 14.0] versus Group BC median 14.0 mm [range 13.1 to 14.5]; difference of medians 1.3 mm; p = 0.02). CONCLUSION: Hemarthrosis damages the synovial tissues and cartilage in the knees of rabbits, regardless of whether they are treated with intra-articular bevacizumab. However, intra-articular injection of bevacizumab may reduce cartilage and synovial damage in rabbits when treatment is initiated early during the development of hemophilic arthropathy. CLINICAL RELEVANCE: If the findings in this study are replicated in larger-animal models that consider the limitations of our work, then a trial in humans might be appropriate to ascertain whether intra-articular injection of bevacizumab could reduce cartilage damage and synovial changes in patients with hemophilia whose hemarthroses cannot otherwise be controlled.


Assuntos
Cartilagem Articular , Osteoartrite , Humanos , Coelhos , Masculino , Animais , Bevacizumab/farmacologia , Bevacizumab/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Hemartrose/tratamento farmacológico , Hemartrose/etiologia , Hemartrose/metabolismo , Membrana Sinovial/diagnóstico por imagem , Cartilagem Articular/diagnóstico por imagem , Injeções Intra-Articulares
3.
Osteoarthritis Cartilage ; 29(4): 471-479, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33307179

RESUMO

Injury to the meniscus is common and frequently leads to the development of post-traumatic osteoarthritis (PTOA). Many times meniscus injuries occur coincident with anterior cruciate ligament (ACL) injuries and lead to a bloody joint effusion. Hemarthrosis, or bleeding into the joint, has been implicated in degeneration of joint tissues. The goal of this review paper is to understand the pathophysiology of blood-induced joint damage, the possible effects of blood on meniscus tissue, and the implications for current meniscus repair techniques that involve the introduction of blood-derived products into the joint. In this review, we illustrate the similarities in the pathophysiology of joint damage due to hemophilic arthropathy (HA) and osteoarthritis (OA). Although numerous studies have revealed the harmful effects of blood on cartilage and synovium, there is currently a gap in knowledge regarding the effects of hemarthrosis on meniscus tissue homeostasis, healing, and the development of PTOA following meniscus injury. Given that many meniscus repair techniques utilize blood-derived and marrow-derived products, it is essential to understand the effects of these factors on meniscus tissue and the whole joint organ to develop improved strategies to promote meniscus tissue repair and prevent PTOA development.


Assuntos
Sangue/metabolismo , Hemartrose/fisiopatologia , Osteoartrite do Joelho/fisiopatologia , Membrana Sinovial/fisiopatologia , Lesões do Menisco Tibial/terapia , Lesões do Ligamento Cruzado Anterior , Reconstrução do Ligamento Cruzado Anterior , Transplante de Medula Óssea/métodos , Hemartrose/etiologia , Hemartrose/metabolismo , Humanos , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/metabolismo , Plasma Rico em Plaquetas , Procedimentos de Cirurgia Plástica/métodos , Transplante de Células-Tronco/métodos , Membrana Sinovial/metabolismo , Lesões do Menisco Tibial/complicações , Lesões do Menisco Tibial/fisiopatologia , Cicatrização
4.
J Thromb Haemost ; 17(11): 1815-1826, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31301687

RESUMO

BACKGROUND: Vascular remodeling associated with hemophilic arthropathy (HA) may contribute to bleed propagation, but the mechanisms remain poorly understood. OBJECTIVES: To explore molecular mechanisms of HA and the effects of hemostasis correction on synovial vascular remodeling after joint injury in hypocoagulable mice. METHODS: Factor VIII (FVIII)-deficient mice +/- FVIII treatment and hypocoagulable wild-type mice (Hypo BALB/c) were subjected to subpatellar puncture. Hypo BALB/c mice were treated with warfarin and anti-FVIII before injury, after which warfarin was continued for 2 weeks or reversed +/- continuous anti-FVIII until harvest. Synovial vascularity was analyzed at baseline and 2 to 4 weeks post injury by histology, musculoskeletal ultrasound with power Doppler (microvascular flow), and Evans blue extravasation (vascular permeability). Synovial gene expression and systemic markers of vascular collagen turnover were studied in FVIII-deficient mice by RNA sequencing and enzyme-linked immunosorbent assay. RESULTS: Vascular changes occurred in FVIII-deficient and Hypo BALB/c mice after injury with minimal effect of hemostasis correction. Increased vascular permeability was only significant in FVIII-deficient mice, who exhibited more pronounced vascular remodeling than Hypo BALB/c mice despite similar bleed volumes. FVIII-deficient mice exhibited a strong transcriptional response in synovium that was only partially affected by FVIII treatment and involved genes relating to angiogenesis and extracellular matrix remodeling, with vascular collagen turnover markers detected systemically. CONCLUSIONS: Intact hemostasis at the time of hemarthrosis and during healing are both critical to prevent vascular remodeling, which appears worse with severe and prolonged FVIII deficiency. Unbiased RNA sequencing revealed potential targets for intervention and biomarker development to improve management of HA.


Assuntos
Permeabilidade Capilar , Fator VIII/metabolismo , Hemartrose/metabolismo , Hemofilia A/metabolismo , Membrana Sinovial/irrigação sanguínea , Remodelação Vascular , Animais , Permeabilidade Capilar/efeitos dos fármacos , Modelos Animais de Doenças , Fator VIII/administração & dosagem , Fator VIII/genética , Feminino , Hemartrose/genética , Hemartrose/fisiopatologia , Hemartrose/prevenção & controle , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemofilia A/fisiopatologia , Hemostasia , Hemostáticos/administração & dosagem , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fatores de Tempo , Remodelação Vascular/efeitos dos fármacos
5.
J Orthop Res ; 37(9): 2043-2052, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31095777

RESUMO

Anterior cruciate ligament reconstructive surgery can restore biomechanical stability, however, such surgery cannot reliably prevent the onset of post-traumatic osteoarthritis. The aim of this study was to elucidate the molecular response that occurs within the menisci following a surgical injury that allows bleeding into the joint space, and then to investigate the effect of dexamethasone (DEX) on this molecular response. Cell viability studies following acute controlled exposure to blood and blood plus DEX were also conducted. Forty-eight New Zealand white rabbits were randomly allocated into control, sham, surgical, and surgical + DEX groups (each group n = 6). Animals were sacrificed at 48 h and 9 weeks, and menisci were harvested. The messenger RNA (mRNA) expression levels for key inflammatory, and degradative proteins, as well as mRNA levels for autophagy pathway molecules were quantified, and statistically significant changes were described. Meniscal cell viability was calculated by incubating groups of medial and lateral menisci in autologous blood, or autologous blood plus DEX for 48 h (each group n = 4; total of eight medial and eight lateral menisci), and then conducting a histological live/dead assay. Results indicated a significant reduction in only medial meniscal cell viability when the tissue was exposed to blood in combination with DEX. A single administration of DEX following surgery significantly suppresses the elevated molecular expression for key inflammatory and degradative markers within menisci at 48 h and 9 weeks post-surgery. In vitro, autologous blood did not affect cell viability, but addition of DEX uniquely impacted the medial menisci. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2043-2052, 2019.


Assuntos
Dexametasona/administração & dosagem , Hemartrose/metabolismo , Meniscos Tibiais/metabolismo , Animais , Autofagia , Sobrevivência Celular/efeitos dos fármacos , Feminino , Hemartrose/patologia , Injeções Intra-Articulares , Metaloproteinase 3 da Matriz/genética , Meniscos Tibiais/patologia , RNA Mensageiro/análise , Coelhos
6.
Blood ; 132(10): 1064-1074, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-29776906

RESUMO

Hemophilic arthropathy (HA) is a debilitating degenerative joint disease that is a major manifestation of the bleeding disorder hemophilia A. HA typically begins with hemophilic synovitis that resembles inflammatory arthritides, such as rheumatoid arthritis, and frequently results in bone loss in patients. A major cause of rheumatoid arthritis is inappropriate release of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) by the TNF-α convertase (TACE; also referred to as ADAM17) and its regulator, iRhom2. Therefore, we hypothesized that iRhom2/ADAM17-dependent shedding of TNF-α also has a pivotal role in mediating HA. Here, we show that addition of blood or its components to macrophages activates iRhom2/ADAM17-dependent TNF-α shedding, providing the premise to study the activation of this pathway by blood in the joint in vivo. For this, we turned to hemophilic FVIII-deficient mice (F8-/- mice), which develop a hemarthrosis following needle puncture injury with synovial inflammation and significant osteopenia adjacent to the affected joint. We found that needle puncture-induced bleeding leads to increased TNF-α levels in the affected joint of F8-/- mice. Moreover, inactivation of TNF-α or iRhom2 in F8-/- mice reduced the osteopenia and synovial inflammation that develops in this mouse model for HA. Taken together, our results suggest that blood entering the joint activates the iRhom2/ADAM17/TNF-α pathway, thereby contributing to osteopenia and synovitis in mice. Therefore, this proinflammatory signaling pathway could emerge as an attractive new target to prevent osteoporosis and joint damage in HA patients.


Assuntos
Proteína ADAM17/metabolismo , Reabsorção Óssea/metabolismo , Proteínas de Transporte/metabolismo , Hemartrose/metabolismo , Hemofilia A/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Proteína ADAM17/genética , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Proteínas de Transporte/genética , Modelos Animais de Doenças , Fator VIII/genética , Feminino , Hemartrose/genética , Hemartrose/patologia , Hemofilia A/genética , Hemofilia A/patologia , Camundongos , Camundongos Knockout , Fator de Necrose Tumoral alfa/genética
7.
Blood ; 129(15): 2161-2171, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28039188

RESUMO

Wound healing requires interactions between coagulation, inflammation, angiogenesis, cellular migration, and proliferation. Healing in dermal wounds of hemophilia B mice is delayed when compared with hemostatically normal wild-type (WT) mice, with abnormal persistence of iron deposition, inflammation, and neovascularity. We observed healing following induced joint hemorrhage in WT and factor IX (FIX) knockout (FIX-/-) mice, examining also parameters previously studied in an excisional skin wound model. Hemostatically normal mice tolerated this joint bleeding challenge, cleared blood from the joint, and healed with minimal pathology, even if additional autologous blood was injected intra-articularly at the time of wounding. Following hemarthrosis, joint wound healing in hemophilia B mice was impaired and demonstrated similar abnormal histologic features as previously described in hemophilic dermal wounds. Therefore, studies of pathophysiology and therapy of hemophilic joint bleeding performed in hemostatically normal animals are not likely to accurately reflect the healing defect of hemophilia. We additionally explored the hypothesis that the use of a FIX replacement protein with extended circulating FIX activity could improve synovial and osteochondral wound healing in hemophilic mice, when compared with treatment with unmodified recombinant FIX (rFIX) in the established joint bleeding model. Significantly improved synovial wound healing and preservation of normal osteochondral architecture are achieved by extending FIX activity after hemarthrosis using glycoPEGylated FIX when compared with an equivalent dose of rFIX. These results suggest that treating joint bleeding only until hemostasis is achieved may not result in optimal joint healing, which is improved by extending factor activity.


Assuntos
Fator IX , Hemartrose , Hemofilia B , Articulações , Pele , Cicatrização , Animais , Modelos Animais de Doenças , Fator IX/genética , Fator IX/farmacologia , Hemartrose/tratamento farmacológico , Hemartrose/genética , Hemartrose/metabolismo , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Hemofilia B/metabolismo , Articulações/lesões , Articulações/metabolismo , Camundongos , Camundongos Knockout , Pele/lesões , Pele/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/genética
8.
Haemophilia ; 22(6): e527-e536, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27704689

RESUMO

INTRODUCTION: Joint haemorrhage is the principal clinical manifestation of haemophilia frequently leading to advanced arthropathy and arthrofibrosis, resulting in severe disability. The degree and prevalence of arthrofibrosis in hemophilic arthropathy is more severe than in other forms of arthropathy. Expression of connective tissue growth factor (CTGF) has been linked to many fibrotic diseases, but has not been studied in the context of haemophilic arthropathy. AIM: We aim to compare synovial tissues histologically from haemophilia and osteoarthritis patients with advanced arthropathy in order to compare expression of proteins that are possibly aetiologic in the development of arthrofibrosis. METHODS: Human synovial tissues were obtained from 10 haemophilia and 10 osteoarthritis patients undergoing joint surgery and processed for histology and immunohistochemistry. RESULTS: All samples from haemophilia patients had synovitis with hypertrophy and hyperplasia of synovial villi. Histologically, synovial tissues contained hyperplastic villi with increased cellularity and abundant haemosiderin- and ferritin-pigmented macrophage-like cells (HMCs), with a perivascular localization in the sub-surface layer. CTGF staining was observed in the surface layer and sub-surface layer in all haemophilia patients, exclusively co-localizing with HMCs. Quantification showed that the extent of CTGF-positive areas was correlated with the degree of detection of HMCs. CTGF was not observed in any of the samples from osteoarthritis patients. CONCLUSION: Using histological analysis, we showed that CTGF expression is elevated in haemophilia patients with arthrofibrosis and absent in patients with osteoarthritis. Additionally, we found that CTGF is always associated with haemosiderin-pigmented macrophage-like cells, which suggests that CTGF is produced by synovial A cells following the uptake of blood breakdown products.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Hemartrose/metabolismo , Hemofilia A/metabolismo , Artropatias/metabolismo , Adulto , Feminino , Hemartrose/complicações , Hemofilia A/complicações , Humanos , Artropatias/etiologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
9.
Am J Sports Med ; 43(11): 2822-32, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26362437

RESUMO

BACKGROUND: Sport injuries of the knee often lead to posttraumatic arthritis. In addition to direct damage of the cartilage, trauma-associated intra-articular bleeding may cause hemarthrosis. Both blood exposure and trauma are known to induce cell death and inflammation and to enhance proteoglycan release in cartilage. HYPOTHESIS: Blood exposure increases chondrocyte death as well as inflammatory and degenerative processes in traumatized cartilage. STUDY DESIGN: Controlled laboratory study. METHODS: Human macroscopically intact osteoarthritic (OA) cartilage explants were impacted by a drop-tower system (0.59 J) and cultivated with or without 10% blood. Interactive effects were studied concerning cell survival, gene expression, and the release of mediators over 24 hours and 96 hours. To evaluate the effects of trauma and hemarthrosis in vivo, a newly established blunt cartilage trauma model in the rabbit was used. Treatment of the knee joints of mature New Zealand White rabbits consisted of the following groups: control (C), arthrotomy (A), arthrotomy with cartilage trauma (AT; 1.0 J), and arthrotomy with cartilage trauma and blood injection (ATH). After 1 and 12 weeks, inflammatory mediators in the synovial fluid and histological changes of the cartilage were determined, and immunohistological staining was performed. RESULTS: The in vitro studies revealed a significant additional or synergistic effect of blood exposure on trauma-induced chondrocyte death, interleukin (IL)-1ß and prostaglandin-E2 (PGE2) release, and matrix metalloproteinase (MMP)/pro-MMP level. Singular arthrotomy in vivo induced a temporary inflammation. Histologically, cartilage trauma caused significant OA changes that were not aggravated by an additional hemarthrosis. Trauma led to a persistent deposition of terminal complement complex (TCC), being enhanced by hemarthrosis. However, trauma-induced formation of osteophytes and arthrotomy-induced elevation of tumor necrosis factor-α release were reduced by hemarthrosis. CONCLUSION: While blood exposure clearly aggravated trauma-induced OA processes in the in vitro model, a singular blood injection revealed heterogeneous effects in vivo, enhancing TCC deposition but reducing trauma-induced osteophyte formation while the histological score of traumatized cartilage was not further impaired. CLINICAL RELEVANCE: The results of this study indicate that a singular, limited bleeding event might not exacerbate early trauma-induced cartilage degeneration in joint injuries. An early removal of intra-articular blood may not prevent the final resulting cartilage damage.


Assuntos
Cartilagem Articular/lesões , Condrócitos/patologia , Inflamação/patologia , Articulação do Joelho/patologia , Idoso , Animais , Morte Celular , Sobrevivência Celular , Dinoprostona/metabolismo , Feminino , Expressão Gênica , Hemartrose/metabolismo , Humanos , Interleucina-1beta/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Pessoa de Meia-Idade , Proteoglicanas/metabolismo , Coelhos , Líquido Sinovial/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Am J Hematol ; 90(11): 1027-35, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26257191

RESUMO

Hemophilic arthropathy is a debilitating condition that can develop as a consequence of frequent joint bleeding despite adequate clotting factor replacement. The mechanisms leading to repeated spontaneous bleeding are unknown. We investigated synovial, vascular, stromal, and cartilage changes in response to a single induced hemarthrosis in the FVIII-deficient mouse. We found soft-tissue hyperproliferation with marked induction of neoangiogenesis and evolving abnormal vascular architecture. While soft-tissue changes were rapidly reversible, abnormal vascularity persisted for months and, surprisingly, was also seen in uninjured joints. Vascular changes in FVIII-deficient mice involved pronounced remodeling with expression of α-Smooth Muscle Actin (SMA), Endoglin (CD105), and vascular endothelial growth factor, as well as alterations of joint perfusion as determined by in vivo imaging. Vascular architecture changes and pronounced expression of α-SMA appeared unique to hemophilia, as these were not found in joint tissue obtained from mouse models of rheumatoid arthritis and osteoarthritis and from patients with the same conditions. Evidence that vascular changes in hemophilia were significantly associated with bleeding and joint deterioration was obtained prospectively by dynamic in vivo imaging with musculoskeletal ultrasound and power Doppler of 156 joints (elbows, knees, and ankles) in a cohort of 26 patients with hemophilia at baseline and during painful episodes. These observations support the hypothesis that vascular remodeling contributes significantly to bleed propagation and development of hemophilic arthropathy. Based on these findings, the development of molecular targets for angiogenesis inhibition may be considered in this disease.


Assuntos
Fator VIII/genética , Hemartrose/patologia , Hemofilia A/patologia , Neovascularização Patológica/patologia , Remodelação Vascular , Actinas/genética , Actinas/metabolismo , Animais , Tornozelo/irrigação sanguínea , Tornozelo/patologia , Modelos Animais de Doenças , Articulação do Cotovelo/irrigação sanguínea , Articulação do Cotovelo/metabolismo , Articulação do Cotovelo/patologia , Endoglina , Fator VIII/metabolismo , Expressão Gênica , Hemartrose/genética , Hemartrose/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Articulação do Joelho/irrigação sanguínea , Articulação do Joelho/metabolismo , Articulação do Joelho/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
J Clin Pathol ; 68(8): 592-600, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25897098

RESUMO

Joint damage due to (recurrent) joint bleeding in haemophilia causes major morbidity. Although the exact pathogenesis has not been fully elucidated, a central role for iron is hypothesised. Likewise, in hereditary haemochromatosis joint destruction is caused by iron overload. A comparison between these types of arthropathy could provide more insight in the influence of iron in inducing joint damage. A literature review was performed to compare both disorders with respect to their clinical and histological characteristics, and preclinical studies on the influence of iron on different joint components were reviewed. Similarities in the features of arthropathy in haemochromatosis and haemophilia are cartilage degeneration, subchondral bone changes with osteophyte and cyst formation, and osteoporosis. In both disorders synovial inflammation and proliferation are seen, although this is much more explicit in haemophilia. Other substantial differences are the age at onset, the occurrence of chondrocalcinosis radiographically and calcium pyrophosphate dihydrate deposition disease in haemochromatosis, and a rapid progression with joint deformity and neovascularisation in haemophilia. Preclinical studies demonstrate detrimental effects of iron to all components of the joint, resulting in synovial inflammation and hyperplasia, chondrocyte death, and impaired osteoblast function. These effects, particularly the synovial changes, are aggravated in the presence of a pro-inflammatory signal, which is prominent in haemophilic arthropathy and minimal in haemochromatosis. Additional research is needed to further specify the role of iron as a specific target in treating these types of arthropathy.


Assuntos
Hemartrose/etiologia , Hemocromatose/complicações , Hemofilia A/complicações , Ferro/metabolismo , Articulações/metabolismo , Animais , Progressão da Doença , Hemartrose/diagnóstico , Hemartrose/metabolismo , Hemocromatose/diagnóstico , Hemocromatose/metabolismo , Hemofilia A/diagnóstico , Hemofilia A/metabolismo , Humanos , Articulações/patologia , Prognóstico , Fatores de Risco
12.
Rev. saúde pública ; 49: 1-9, 27/02/2015. tab, graf
Artigo em Inglês | LILACS | ID: lil-742296

RESUMO

OBJECTIVE To validate a screening instrument using self-reported assessment of frailty syndrome in older adults. METHODS This cross-sectional study used data from the Saúde, Bem-estar e Envelhecimento study conducted in Sao Paulo, SP, Southeastern Brazil. The sample consisted of 433 older adult individuals (≥ 75 years) assessed in 2009. The self-reported instrument can be applied to older adults or their proxy respondents and consists of dichotomous questions directly related to each component of the frailty phenotype, which is considered the gold standard model: unintentional weight loss, fatigue, low physical activity, decreased physical strength, and decreased walking speed. The same classification proposed in the phenotype was utilized: not frail (no component identified); pre-frail (presence of one or two components), and frail (presence of three or more components). Because this is a screening instrument, “process of frailty” was included as a category (pre-frail and frail). Cronbach’s α was used in psychometric analysis to evaluate the reliability and validity of the criterion, the sensitivity, the specificity, as well as positive and negative predictive values. Factor analysis was used to assess the suitability of the proposed number of components. RESULTS Decreased walking speed and decreased physical strength showed good internal consistency (α = 0.77 and 0.72, respectively); however, low physical activity was less satisfactory (α = 0.63). The sensitivity and specificity for identifying pre-frail individuals were 89.7% and 24.3%, respectively, while those for identifying frail individuals were 63.2% and 71.6%, respectively. In addition, 89.7% of the individuals from both the evaluations were identified in the “process of frailty” category. CONCLUSIONS The self-reported assessment of frailty can identify the syndrome among older adults and can be used as a screening tool. Its ...


OBJETIVO Validar instrumento de rastreamento por avaliação autorreferida da síndrome de fragilidade entre idosos. MÉTODOS Estudo transversal com dados do estudo Saúde, Bem-estar e Envelhecimento, realizado em São Paulo, SP. A amostra probabilística foi constituída por 433 idosos (idade ≥ 75 anos) avaliados em 2009. O instrumento autorreferido utilizado pode ser aplicado a idosos ou proxi-informantes e foi composto por questões dicotômicas relacionadas diretamente a cada componente do fenótipo de fragilidade considerado padrão-ouro: perda de peso não intencional, fadiga, baixa atividade física, redução de força e de velocidade de marcha. Manteve-se a classificação proposta no fenótipo: não frágil (nenhum componente identificado); pré-frágil (presença de um ou dois componentes) e frágil (presença de três ou mais componentes). Por tratar-se de instrumento de rastreamento, incluiu-se a categoria processo de fragilização (pré-frágil e frágil). Utilizou-se o coeficiente α de Cronbach na análise psicométrica para avaliar confiabilidade e validade de critério, sensibilidade, especificidade e valores preditivos positivo e negativo. Para verificar a adequação do número de componentes propostos, utilizou-se a análise fatorial. RESULTADOS Os componentes “redução de velocidade de caminhada” e “redução de força” apresentaram boa consistência interna (α = 0,77 e α = 0,72, respectivamente) e a “baixa atividade física” (α = 0,63) foi um pouco menos satisfatória. A sensibilidade e a especificidade para identificação dos pré-frágeis foram de 89,7% e 24,3% e dos frágeis, 63,2% e 71,6%, respectivamente. A categoria “processo de fragilização” identificou, igualmente, 89,7% das pessoas em ambas as avaliações. CONCLUSÕES O instrumento de avaliação de fragilidade autorreferida é capaz de identificar a síndrome entre as pessoas idosas, podendo ser utilizado como instrumento de rastreamento, ...


Assuntos
Animais , Humanos , Proteínas ADAM/metabolismo , Proteínas de Transporte/metabolismo , Hemartrose/etiologia , Hemartrose/metabolismo , Hemofilia A/complicações , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Sinovite/etiologia , Sinovite/metabolismo
13.
Haemophilia ; 20 Suppl 4: 11-4, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24762269

RESUMO

One of the main complications of haemophilia A is haemophilic arthropathy (HA), a debilitating disease with a significant negative impact on motility and quality of life. Despite major advances in the treatment of haemophilia A, many patients still suffer from HA. We wish to develop new treatments for HA, but must first better understand its causes. Our laboratory studies molecular scissors that release the pro-inflammatory cytokine tumour necrosis factor alpha (TNFα) from cells. TNFα is considered the 'fire alarm' of the body - it helps to fight infections, but can also cause diseases such as inflammatory arthritis. We know that the molecular scissors, called TNFα convertase (TACE), and its newly discovered regulator termed iRhom2 can be rapidly activated by small amounts of cytokines, growth factors, and pro-inflammatory mediators present in the blood. We hypothesize that the rapid activation of TACE could help explain one of the unsolved mysteries regarding the development of HA, which is how even small amounts of blood can provoke a persistent inflammatory response. We propose that once blood enters the joint, iRhom2 and TACE are activated to release TNFα and that this could promote the development of HA in a similar manner to that in which it promotes rheumatoid arthritis (RA). We are currently using immune cells stimulated with blood degradation products, and mouse models of HA, to test this hypothesis. If successful, our study could provide the rationale for testing anti-TNF antibodies, which are already used to treat RA, for the treatment of HA. In addition, they might uncover iRhom2 and TACE as attractive new candidate targets for the treatment of HA.


Assuntos
Proteínas ADAM/metabolismo , Proteínas de Transporte/metabolismo , Hemartrose/etiologia , Hemartrose/metabolismo , Hemofilia A/complicações , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Proteína ADAM17 , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Sinovite/etiologia , Sinovite/metabolismo
14.
Am J Sports Med ; 42(5): 1096-102, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24664137

RESUMO

BACKGROUND: Osteochondral fractures are often seen on magnetic resonance imaging (MRI) of acutely injured knees, but their existence has gained little interest because of a lack of knowledge of their relation to treatment options and outcome. It is not clear whether acute phase synovial fluid (SF) concentrations of cartilage and bone markers and proinflammatory cytokines are different between traumatically injured knees with or without osteochondral fracture. HYPOTHESIS: Acutely injured knees with an osteochondral fracture, particularly fractures with disrupted cortical bone, have higher concentrations of bone markers and cytokines than do knees without an osteochondral fracture. STUDY DESIGN: Cross-sectional study; Level of evidence, 3. METHODS: Synovial fluid (hemarthrosis) was aspirated (median 1 day after injury) and 1.5-T MRI was performed (median 8 days after injury) in the acutely injured knee of 98 individuals (26% women; mean age, 23 years). As visualized on MRI, 39% knees had an osteochondral fracture with disrupted cortical bone, 30% had an osteochondral fracture with intact cortical bone, and 32% did not have an osteochondral fracture. Concentrations of sulfated glycosaminoglycan, ARGS aggrecan, cartilage oligomeric matrix protein, osteocalcin, secreted protein acidic and rich in cysteine (SPARC), osteopontin and proinflammatory cytokines (interleukin [IL]-1ß, IL-6, IL-8, and tumor necrosis factor [TNF]-α) were analyzed. RESULTS: After adjusting for days between injury and SF aspiration, age at injury, and sex, knees with any osteochondral fracture (with or without disrupted cortical bone) had significantly higher SF concentrations of TNF-α (median [interquartile range (IQR)] = 9 [7-12] pg/mL vs. 7 [5-14] pg/mL; P = .013), whereas knees with an osteochondral fracture with disrupted cortical bone had significantly higher SF concentrations (medians [IQRs]) of SPARC (492 [328-754] ng/mL vs. 407 [140-685] ng/mL; P = .030), IL-8 (278 [148-628] pg/mL vs. 138 [67-413] pg/mL; P = .028), and TNF-α (11 [7-15] pg/mL vs. 7 [5-14] pg/mL; P = .004) compared with knees without an osteochondral fracture. CONCLUSION: In acutely injured knees with hemarthrosis, a concomitant osteochondral fracture with disrupted cortical bone is associated with a higher degree of joint inflammation.


Assuntos
Artrite/metabolismo , Fraturas Ósseas/complicações , Fraturas de Cartilagem/complicações , Traumatismos do Joelho/complicações , Lesões dos Tecidos Moles/complicações , Líquido Sinovial/metabolismo , Doença Aguda , Adolescente , Adulto , Biomarcadores/metabolismo , Proteína de Matriz Oligomérica de Cartilagem/metabolismo , Cartilagem Articular/lesões , Estudos Transversais , Feminino , Fraturas Ósseas/diagnóstico , Fraturas de Cartilagem/diagnóstico , Hemartrose/metabolismo , Humanos , Interleucina-8/metabolismo , Traumatismos do Joelho/diagnóstico , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Osteonectina/metabolismo , Luxação Patelar/complicações , Luxação Patelar/diagnóstico , Ligamento Cruzado Posterior/lesões , Lesões dos Tecidos Moles/diagnóstico , Sucção , Lesões do Menisco Tibial , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
15.
Haemophilia ; 20(1): e32-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24308756

RESUMO

Haemophilic arthropathy (HA) is characterized by chronic proliferative synovitis leading to cartilage destruction and shares some pathological features with rheumatoid arthritis (RA). Apoptosis has been implicated in RA pathogenesis, and an agonistic anti-Fas monoclonal antibody (mAb) was found to induce RA fibroblast-like synoviocyte (FLS) apoptosis and suppress synovial hyperplasia in animal models of RA. The aim of this study was to evaluate the effect of anti-Fas mAb on HA-FLS. FLS were isolated from knee synovial biopsies from six HA patients, six RA patients and six healthy subjects. The expression of Fas in synovial biopsies was investigated by immunohistochemistry. FLS were stimulated with anti-Fas mAb at different concentrations, alone or in combination with tumour necrosis factor-α (TNF-α) and basic fibroblast growth factor (bFGF). Fas expression in FLS was assessed by Western blot. Cell viability was studied with the WST-1 assay. Active caspase-3 levels were measured using ELISA and Western blot. A strong Fas-immunoreactivity was observed in different cells of HA synovium, including FLS, inflammatory cells and endothelial cells. Fas antigen was constitutively overexpressed in cultured HA-FLS. Anti-Fas mAb had a significant cytotoxicity on HA-FLS in a dose-dependent manner, either alone or in combination with TNF-α and bFGF. These cytotoxic effects were due to the ability of anti-Fas to induce HA-FLS apoptosis, as shown by the increased active caspase-3 levels. Anti-Fas mAb exhibited a more pronounced pro-apoptotic effect on HA-FLS than RA-FLS. Fas antigen is highly expressed on HA-FLS and its stimulation by anti-Fas mAb may be an effective strategy to induce HA-FLS apoptosis.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Hemartrose/etiologia , Hemofilia A/complicações , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/patologia , Adulto , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Murinos , Artrite Reumatoide/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/metabolismo , Hemartrose/metabolismo , Hemartrose/patologia , Humanos , Imunoglobulina M/imunologia , Imunoglobulina M/farmacologia , Masculino , Pessoa de Meia-Idade , Membrana Sinovial/metabolismo , Adulto Jovem , Receptor fas/imunologia , Receptor fas/metabolismo
16.
Haemophilia ; 20(3): 421-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24330418

RESUMO

The penetration of beta energy of 153-samarium ((153) Sm) (0.8 MeV) is not only appropriate for synovectomy of median articulations but is possible to improve the radiobiological effect using increased activities. The aim of this study was to assess the effectiveness of 185 MBq and 740 MBq of 153-samarium hydroxyapatite ((153) Sm-HA) in knees of haemophilic patients. Thirty-one patients--36 knees, 30 males, were divided into two groups without coinjection of corticosteroid: A - 14 patients (17 knees) treated with intra-articular dose of 185 MBq of (153) Sm-HA, average age 23 years; B--17 patients (19 knees) with 740 MBq of (153) Sm-HA, average age 21.3 years. The evaluation before and after 1 year of synovectomy used the following criteria: reduction in the number of haemarthroses and use of the coagulation factor and improvement in articular motility. Adverse-effects occurrence was considered too. Early and late scintigraphic studies were performed after synoviorthesis and no joint immobilization was recommended. The reduction in haemarthrosis and use of coagulation factor were: group 1--31.3% and 25%; group 2--81.5% and 79% with P < 0.001 respectively; no significant improvement in knees motility was noted for both groups. Four cases of mild reactional synovitis were observed in each group. The scintigraphic control showed homogenous distribution of the radiopharmaceuticals with no articular escape; the material was considered safe by its permanence in the articulation. We have significant improvement in the synovectomy of haemophilic knees with 740 MBq of (153) Sm-HA; the less penetration of its beta radiation was compensated by the increased biological effect with the higher used activity.


Assuntos
Hemartrose/radioterapia , Hemofilia A/complicações , Hidroxiapatitas/administração & dosagem , Radioisótopos/administração & dosagem , Samário/administração & dosagem , Sinovite/etiologia , Sinovite/radioterapia , Adolescente , Criança , Relação Dose-Resposta à Radiação , Feminino , Hemartrose/etiologia , Hemartrose/metabolismo , Humanos , Hidroxiapatitas/farmacocinética , Injeções Intra-Articulares , Articulação do Joelho/metabolismo , Articulação do Joelho/fisiopatologia , Articulação do Joelho/efeitos da radiação , Masculino , Estudos Prospectivos , Samário/farmacocinética , Sinovite/metabolismo , Resultado do Tratamento , Adulto Jovem
17.
Haemophilia ; 19(4): e218-27, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23777533

RESUMO

Recurrent joint bleeding is the most common manifestation of severe haemophilia resulting in haemophilic arthropathy (HA). Iron plays a central role in the pathogenesis of the two main features of HA: synovitis and cartilage destruction. The aim of this study was to investigate the synovial presence of the iron regulator proteins ferroportin (FPN), hepcidin, haemoglobin scavenger receptor CD163 (CD163), feline leukaemia virus subgroup C (FLVCR), and heme carrier protein 1 (HCP-1). A comparison of the expression in HA with rheumatoid arthritis (RA), osteoarthritis (OA), and healthy controls (HC) is made. Synovial expression of iron regulators was investigated by immunohistochemistry in human synovial tissue and in a murine haemophilia model. We demonstrate for the first time the synovial presence of the investigated iron regulator proteins. Expression of the iron regulator proteins FPN, CD163, FLVCR, and HCP-1 was enhanced in HA in comparison to RA, OA, and HC synovium. In addition, in a murine haemophilia model of acute joint bleeding, synovial expression of FPN, CD163, and HCP-1 was increased. In both human and murine experiment, synovial expression of hepcidin was not altered. These findings indicate the presence of iron regulator proteins in the synovium, demonstrate an enhanced expression of FPN, CD163, FLVCR, and HCP-1 in HA, and suggest a synovial adaptation mechanism to maintain synovial iron homeostasis in HA.


Assuntos
Artrite Reumatoide/metabolismo , Hemartrose/metabolismo , Hemofilia A/metabolismo , Proteínas Reguladoras de Ferro/metabolismo , Osteoartrite/metabolismo , Membrana Sinovial/metabolismo , Regulação para Cima , Animais , Artrite Reumatoide/patologia , Estudos de Casos e Controles , Modelos Animais de Doenças , Hemartrose/patologia , Hemofilia A/patologia , Humanos , Ferro/metabolismo , Camundongos , Osteoartrite/patologia , Membrana Sinovial/patologia
18.
Rheumatology (Oxford) ; 52(9): 1563-71, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23418047

RESUMO

OBJECTIVE: IL-4 plus IL-10 prevents blood-induced cartilage damage. The aim of the present study was to evaluate whether cartilage damage can still be averted by addition of IL-4 plus IL-10 when added after the onset of a bleed and whether aspiration of blood prior to addition of IL-4 plus IL-10 is of additive protective value. METHODS: Healthy canine hip and human shoulder cartilage was exposed to whole blood for 4 days. IL-4 plus IL-10 was administered directly or after a delay of several hours up to 2 days. Furthermore, blood was aspirated after 1 or 2 days and subsequently IL-4 plus IL-10 was added. IL-1ß concentration and cartilage matrix proteoglycan turnover were determined. RESULTS: Exposure of canine and human cartilage to blood decreased the proteoglycan synthesis rate and content and increased proteoglycan release. IL-4 plus IL-10 only prevented blood-induced damage of canine cartilage when added directly, not after 4 h or later. For human cartilage, IL-4 plus IL-10 limited blood-induced damage as well as IL-1ß production when administered within 4-8 h after the onset of a bleed, but not thereafter. Aspiration of blood within 24 h fully prevented cartilage damage. Subsequent addition of IL-4 plus IL-10 was not of additive value. CONCLUSION: For humans, there is a short time window after onset of a joint bleed in which IL-4 plus IL-10 can limit blood-induced cartilage damage. Furthermore, aspiration of a joint to shorten blood exposure fully prevents cartilage damage. Both options can be considered in the treatment of a joint haemorrhage.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Hemartrose/tratamento farmacológico , Interleucina-10/uso terapêutico , Interleucina-4/uso terapêutico , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Cães , Hemartrose/metabolismo , Hemartrose/patologia , Humanos , Interleucina-1beta/metabolismo
19.
Osteoarthritis Cartilage ; 20(11): 1302-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22874525

RESUMO

PURPOSE: The aim of this cross-sectional study was to investigate concentrations of cartilage and bone markers, and pro-inflammatory cytokines in synovial fluid (SF) collected at different time-points from acutely injured knees with hemarthrosis and to compare these with SF concentrations of knees of age and gender-matched healthy reference subjects. METHODS: SF was aspirated from the acutely injured knee of 111 individuals (mean age 27 years, span 13-64 years, 22% women). Concentrations of sulfated glycosaminoglycan (sGAG) were measured by Alcian blue precipitation whereas cartilage ARGS, bone biomarkers [osteocalcin (OCL), secreted protein acidic and rich in cysteine (SPARC) and osteopontin (OPN)] and pro-inflammatory cytokines [interleukin (IL)-1ß, IL-6, IL-8 and tumor necrosis factor (TNF)-α] were analyzed using electrochemiluminescence. Samples were also analyzed with regard to time between injury and aspiration [same day (n = 29), 1 day (n = 31), 2-3 days (n = 19), 4-7 days (n = 20) and 8-23 days (n = 12)]. RESULTS: SF concentrations of ARGS (P < 0.001), SPARC (P < 0.001), OPN (P < 0.001), and all cytokines (P < 0.001), but not sGAG (P = 0.06) or OCL (P = 0.992), were significantly higher in injured knees compared to knees of reference subjects. The cartilage markers sGAG and ARGS were significantly higher in knees aspirated later than 1 day after injury, whereas concentrations of SPARC and OPN and all cytokines were higher in knees aspirated the same day as the injury and at all time-points thereafter. CONCLUSIONS: Our results suggest that an acute knee injury is associated with an instant local biochemical response to the trauma, which may affect cartilage and bone as well as the inflammatory activity.


Assuntos
Osso e Ossos/metabolismo , Cartilagem Articular/metabolismo , Citocinas/metabolismo , Hemartrose/metabolismo , Traumatismos do Joelho/metabolismo , Líquido Sinovial/metabolismo , Doença Aguda , Adolescente , Adulto , Agrecanas/metabolismo , Biomarcadores/metabolismo , Estudos Transversais , Feminino , Hemartrose/etiologia , Humanos , Traumatismos do Joelho/complicações , Masculino , Pessoa de Meia-Idade , Fatores de Tempo , Adulto Jovem
20.
J Rheumatol ; 39(8): 1678-86, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22753650

RESUMO

OBJECTIVE: Hemarthrosis triggers hemophilic arthropathy, involving the target joints. The histopathogenesis of blood-induced joint damage remains unclear. The triad of receptor activator of nuclear factor-κB (RANK), RANK ligand (RANKL), and osteoprotegerin (OPG; RANK-RANKL-OPG) controls bone turnover. Our aim was to evaluate RANK-RANKL-OPG expression in the synovium of hemophilic patients with severe arthropathy. METHODS: Synovial biopsies were obtained from 18 patients with hemophilic arthropathy and 16 with osteoarthritis (OA) who were undergoing total knee replacement and synovectomy. The severity of hemophilic arthropathy was evaluated according to ultrasonography score, the World Federation of Hemophilia (WFH) orthopedic joint scale, and the radiographic Pettersson score. RANK-RANKL-OPG expression was examined by immunohistochemistry and Western blotting. Serum levels of soluble RANKL (sRANKL) and OPG from an extended group of 67 patients with hemophilic arthropathy and 30 healthy controls were measured by ELISA. RESULTS: The mean ultrasonography, WFH orthopedic joint scale, and Pettersson scores in patients with hemophilic arthropathy indicated severe arthropathy. A decreased expression of OPG was found in hemophilic arthropathy synovium compared with patients with OA. RANK and RANKL immunopositivity was strong in the lining and sublining layers in hemophilic arthropathy synovial tissue. Western blotting confirmed the immunohistological findings. Serum levels of sRANKL and OPG in patients with hemophilia were lower than in healthy controls. CONCLUSION: In hemophilic arthropathy, the synovium highly expressed RANK and RANKL, whereas OPG immunopositivity decreased, suggesting an osteoclastic activation. Low tissue expression of OPG paralleled the serum levels of this protein and the severity of hemophilic arthropathy assessed by ultrasonography, Pettersson, and WFH orthopedic joint scale scores.


Assuntos
Hemartrose/metabolismo , Articulação do Joelho/metabolismo , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Feminino , Hemartrose/diagnóstico por imagem , Hemartrose/patologia , Humanos , Articulação do Joelho/diagnóstico por imagem , Articulação do Joelho/patologia , Masculino , Pessoa de Meia-Idade , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Índice de Gravidade de Doença , Transdução de Sinais/fisiologia , Membrana Sinovial/diagnóstico por imagem , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA