Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Signal Transduct Target Ther ; 9(1): 109, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38714712

RESUMEN

The knee joint has long been considered a closed system. The pathological effects of joint diseases on distant organs have not been investigated. Herein, our clinical data showed that post-traumatic joint damage, combined with joint bleeding (hemarthrosis), exhibits a worse liver function compared with healthy control. With mouse model, hemarthrosis induces both cartilage degeneration and remote liver damage. Next, we found that hemarthrosis induces the upregulation in ratio and differentiation towards Th17 cells of CD4+ T cells in peripheral blood and spleen. Deletion of CD4+ T cells reverses hemarthrosis-induced liver damage. Degeneration of cartilage matrix induced by hemarthrosis upregulates serological type II collagen (COL II), which activates CD4+ T cells. Systemic application of a COL II antibody blocks the activation. Furthermore, bulk RNAseq and single-cell qPCR analysis revealed that the cartilage Akt pathway is inhibited by blood treatment. Intra-articular application of Akt activator blocks the cartilage degeneration and thus protects against the liver impairment in mouse and pig models. Taken together, our study revealed a pathological joint-liver axis mediated by matrikine-activated CD4+ T cells, which refreshes the organ-crosstalk axis and provides a new treatment target for hemarthrosis-related disease. Intra-articular bleeding induces cartilage degradation through down-reulation of cartilage Akt pathway. During this process, the soluble COL II released from the damaged cartilage can activate peripheral CD4+ T cells, differention into Th17 cells and secretion of IL-17, which consequently induces liver impairment. Intra-articular application of sc79 (inhibitor of Akt pathway) can prevent the cartilage damage as well as its peripheral influences.


Asunto(s)
Linfocitos T CD4-Positivos , Hígado , Animales , Ratones , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Hígado/patología , Hígado/metabolismo , Hemartrosis/genética , Hemartrosis/patología , Masculino , Modelos Animales de Enfermedad , Células Th17/inmunología , Células Th17/patología , Colágeno Tipo II/genética , Venenos Elapídicos/farmacología , Femenino , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Mol Genet Genomic Med ; 10(6): e1931, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35426265

RESUMEN

BACKGROUND: Venous malformations (VMs) are the most common vascular anomalies and have been associated with somatic variants in TEK. Current treatment of VM joint component might be challenging due to the size or location of some lesions or ineffective with recurrence of malformed veins. Targeted molecular therapies after identification of genetic defects might be an alternative. METHODS: We report a case with intraarticular bleeding due to VM with a TEK pathogenic somatic variant treated with rapamycin. RESULTS: A 26-year-old female patient was evaluated for right calf pain secondary to venous malformation of the right inferior limb with an intraarticular component in the right knee. Hemarthrosis and degenerative chondropathy of the knee were evidenced at MRA. Molecular diagnosis evidenced a pathogenic somatic TEK variant. Rapamycin was introduced to stop bleeding, with good tolerance and efficacy. CONCLUSION: The TEK receptor signals through the PI3K/AKT/mTOR pathway and TEK mutations have been linked to AKT activation. As rapamycin acts against angiogenesis and reduces phosphorylated-AKT levels, targeted molecular therapy should be discussed as first-line therapy in patients with proven molecular diagnosis and diffuse VM inaccessible to conventional treatment.


Asunto(s)
Enfermedades Vasculares , Malformaciones Vasculares , Adulto , Femenino , Hemartrosis/tratamiento farmacológico , Hemartrosis/genética , Humanos , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt , Sirolimus/uso terapéutico , Malformaciones Vasculares/complicaciones , Malformaciones Vasculares/tratamiento farmacológico , Malformaciones Vasculares/genética
3.
Mol Biol Rep ; 48(1): 969-974, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33289909

RESUMEN

Haemophilic arthropathy (HA), caused by intra-articular haemorrhage, is one of the most common complications in patients with haemophilia. Factor replacement therapy provides missing coagulation factors to prevent children with haemophilia from joint bleeding and decreases their risk for HA. However, haemophilia patients in developing countries are still suffering from HA due to insufficient replacement therapy. Symptoms such as pain and activity limitations caused by HA seriously affect the functional abilities and quality of life of patients with HA, causing a high disability rate in the haemophilia cohort. The pathological mechanism of HA is complicated because the whole pathological mainly involves hypertrophic synovitis, osteopenia, cartilage and bone destruction, and these pathological changes occur in parallel and interact with each other. Inflammation plays an important role in the whole complex pathological process, and iron, cytokines, growth factors and other factors are involved. This review summarizes the pathological mechanism of HA to provide background for clinical and basic research.


Asunto(s)
Artritis/patología , Enfermedades Óseas Metabólicas/patología , Hemartrosis/patología , Hemofilia A/patología , Osteonecrosis/patología , Sinovitis/patología , Adulto , Artritis/genética , Artritis/inmunología , Artritis/metabolismo , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/inmunología , Enfermedades Óseas Metabólicas/metabolismo , Niño , Citocinas/genética , Citocinas/inmunología , Factor VIII/uso terapéutico , Regulación de la Expresión Génica , Hemartrosis/genética , Hemartrosis/inmunología , Hemartrosis/metabolismo , Hemofilia A/genética , Hemofilia A/inmunología , Hemofilia A/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Hierro/inmunología , Hierro/metabolismo , Articulaciones/inmunología , Articulaciones/metabolismo , Articulaciones/patología , Osteonecrosis/genética , Osteonecrosis/inmunología , Osteonecrosis/metabolismo , Calidad de Vida , Sinovitis/genética , Sinovitis/inmunología , Sinovitis/metabolismo
4.
Int J Mol Sci ; 21(19)2020 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023246

RESUMEN

BACKGROUND: Hemophilia A and B are X-linked congenital bleeding disorders characterized by recurrent hemarthroses leading to specific changes in the synovium and cartilage, which finally result in the destruction of the joint: this process is called hemophilic arthropathy (HA). This review highlights the most prominent molecular biomarkers found in the literature to discuss their potential use in the clinical practice to monitor bleeding, to assess the progression of the HA and the effectiveness of treatments. METHODS: A review of the literature was performed on PubMed and Embase, from 3 to 7 August 2020. Study selection and data extraction were achieved independently by two authors and the following inclusion criteria were determined a priori: English language, available full text and articles published in peer-reviewed journal. In addition, further articles were identified by checking the bibliography of relevant articles and searching for the studies cited in all the articles examined. RESULTS: Eligible studies obtained at the end of the search and screen process were seventy-three (73). CONCLUSIONS: Despite the surge of interest in the clinical use of biomarkers, current literature underlines the lack of their standardization and their potential use in the clinical practice preserving the role of physical examination and imaging in early diagnosis.


Asunto(s)
Biomarcadores/sangre , Hemofilia A/sangre , Hemofilia B/sangre , Artropatías/sangre , Genes Ligados a X/genética , Hemartrosis/sangre , Hemartrosis/genética , Hemartrosis/patología , Hemofilia A/genética , Hemofilia A/patología , Hemofilia B/genética , Hemofilia B/patología , Hemorragia/sangre , Hemorragia/patología , Humanos , Artropatías/genética , Artropatías/patología , Membrana Sinovial/patología
5.
Mol Genet Genomic Med ; 7(11): e979, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31566926

RESUMEN

BACKGROUND: The pathophysiology of hemophilic arthropathy is complex and not completely understood. In this study, we aimed to identify biomarkers that can affect the hemophilic arthropathy severity. METHODS: Fifty patients were analyzed for biomarker frequencies; in 37 patients, articular symptoms were evaluated based on the physical joint examination score, and in 18, it was based on magnetic resonance imaging. Eight polymorphisms, namely FV 1691G>A, FII 20210G>A, MTHFR 677C>T and 1298A>C, TNFα-308G>A and -238G>A, ACAN VNTR, and IL1RN*2-VNTR were identified. RESULTS: Patients with the MTHFR 677TT genotype showed a higher number of affected joints (1.83 ± 0.9 vs. 0.55 ± 0.7 for CC; p = .023), whereas those with the MTHFR 1298AC genotype exhibited higher effusion according to two radiologists (0.90 ± 0.31/1.20 ± 0.63 vs. 0.38 ± 0.52/0.50 ± 0.53 for AA genotype; p = .043/0.036, respectively). In addition, patients with the TNFα-308GA genotype had more subchondral cysts (0.75 ± 0.95 vs. 0.07 ± 0.26 for GG genotype; p = .041). CONCLUSIONS: The distribution of risk genotypes for MTHFR and TNFα-308GA suggests their association with clinical parameters of hemophilic arthropathy. Cohort studies are essential to verify these associations.


Asunto(s)
Cartílago/patología , Marcadores Genéticos , Hemartrosis/diagnóstico , Hemofilia A/fisiopatología , Inflamación/diagnóstico , Adolescente , Cartílago/metabolismo , Niño , Preescolar , Femenino , Hemartrosis/epidemiología , Hemartrosis/genética , Humanos , Incidencia , Lactante , Recién Nacido , Inflamación/epidemiología , Inflamación/genética , Masculino , México/epidemiología , Pronóstico
6.
J Thromb Haemost ; 17(11): 1815-1826, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31301687

RESUMEN

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.


Asunto(s)
Permeabilidad Capilar , Factor VIII/metabolismo , Hemartrosis/metabolismo , Hemofilia A/metabolismo , Membrana Sinovial/irrigación sanguínea , Remodelación Vascular , Animales , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Factor VIII/genética , Femenino , Hemartrosis/genética , Hemartrosis/fisiopatología , Hemartrosis/prevención & control , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemofilia A/fisiopatología , Hemostasis , Hemostáticos/administración & dosificación , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Factores de Tiempo , Remodelación Vascular/efectos de los fármacos
7.
J Thromb Haemost ; 17(8): 1240-1246, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31148392

RESUMEN

BACKGROUND: Following induced joint hemorrhage, hemophilia B results in the abnormal persistence of iron deposition, inflammation, and neovascularity of the synovial tissue, as well as deterioration of the bone articular surface and strength. Previously, we demonstrated that a factor IX (FIX) replacement protein with extended circulating FIX activity, glycoPEGylated FIX nonacog beta pegol (N9-GP), could improve synovial and osteochondral parameters in F9 knockout mice when administered after joint injury. OBJECTIVE: We explored the use of N9-GP prior to unilateral joint hemorrhage and compared to unmodified recombinant FIX (rFIX). METHODS: Pharmacodynamics, histology, and microcomputed tomography were used to assess the effects of prophylactic administration of glycoPEGylated FIX. RESULTS: In comparison to rFIX, N9-GP significantly improved soft tissue histological parameters, as well as bone outcome at 2 weeks post injury, while performing equally in reduction of blood present in the joint space assessed 1 day after injury. CONCLUSIONS: These results indicate that, in comparison to rFIX, the prophylactic use of extended half-life FIX provides superior protection from bleeding-induced joint damage, manifested by improved correction of histologic parameters.


Asunto(s)
Factor IX/metabolismo , Hemartrosis/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Hemostáticos/administración & dosificación , Articulaciones/efectos de los fármacos , Polietilenglicoles/administración & dosificación , Animales , Modelos Animales de Enfermedad , Esquema de Medicación , Factor IX/administración & dosificación , Factor IX/genética , Factor IX/farmacocinética , Semivida , Hemartrosis/diagnóstico por imagen , Hemartrosis/genética , Hemartrosis/metabolismo , Hemofilia B/genética , Hemofilia B/metabolismo , Hemostáticos/farmacocinética , Articulaciones/diagnóstico por imagen , Articulaciones/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Polietilenglicoles/farmacocinética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética
8.
Blood ; 132(15): 1593-1603, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30026184

RESUMEN

Joint bleeds are common in congenital hemophilia but rare in acquired hemophilia A (aHA) for reasons unknown. To identify key mechanisms responsible for joint-specific bleeding in congenital hemophilia, bleeding phenotypes after joint injury and tail transection were compared in aHA wild-type (WT) mice (receiving an anti-factor VIII [FVIII] antibody) and congenital HA (FVIII-/-) mice. Both aHA and FVIII-/- mice bled severely after tail transection, but consistent with clinical findings, joint bleeding was notably milder in aHA compared with FVIII-/- mice. Focus was directed to thrombin-activatable fibrinolysis inhibitor (TAFI) to determine its potentially protective effect on joint bleeding in aHA. Joint bleeding in TAFI-/- mice with anti-FVIII antibody was increased, compared with WT aHA mice, and became indistinguishable from joint bleeding in FVIII-/- mice. Measurements of circulating TAFI zymogen consumption after joint injury indicated severely defective TAFI activation in FVIII-/- mice in vivo, consistent with previous in vitro analyses in FVIII-deficient plasma. In contrast, notable TAFI activation was observed in aHA mice, suggesting that TAFI protected aHA joints against bleeding. Pharmacological inhibitors of fibrinolysis revealed that urokinase-type plasminogen activator (uPA)-induced fibrinolysis drove joint bleeding, whereas tissue-type plasminogen activator-mediated fibrinolysis contributed to tail bleeding. These data identify TAFI as an important modifier of hemophilic joint bleeding in aHA by inhibiting uPA-mediated fibrinolysis. Moreover, our data suggest that bleed protection by TAFI was absent in congenital FVIII-/- mice because of severely defective TAFI activation, underscoring the importance of clot protection in addition to clot formation when considering prohemostatic strategies for hemophilic joint bleeding.


Asunto(s)
Carboxipeptidasa B2/metabolismo , Hemartrosis/etiología , Hemartrosis/metabolismo , Hemofilia A/complicaciones , Animales , Carboxipeptidasa B2/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Hemartrosis/genética , Hemofilia A/genética , Hemofilia A/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
9.
Blood ; 132(10): 1064-1074, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29776906

RESUMEN

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.


Asunto(s)
Proteína ADAM17/metabolismo , Resorción Ósea/metabolismo , Proteínas Portadoras/metabolismo , Hemartrosis/metabolismo , Hemofilia A/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteína ADAM17/genética , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Factor VIII/genética , Femenino , Hemartrosis/genética , Hemartrosis/patología , Hemofilia A/genética , Hemofilia A/patología , Ratones , Ratones Noqueados , Factor de Necrosis Tumoral alfa/genética
10.
Thromb Haemost ; 117(11): 2092-2104, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29202211

RESUMEN

Background Replacement therapy with coagulation factor VIII (FVIII) concurrent with bleeds (on-demand) in haemophilia A (HA) patients has been hypothesized to increase the risk for antidrug antibodies (inhibitors). A danger signal environment, characterized by tissue damage and inflammation at the site of a bleed, is thought to contribute to the anti-FVIII response. The nature of this inflammatory reaction is, however, not fully known, and new insights will be valuable for both managing inhibitors and understanding arthropathy development. Objective To characterize the inflammatory response, locally and systemically, during the first 24 hours following a joint bleed in the HA rat. Methods HA rats received a needle-induced knee joint bleed (n = 83) or a sham procedure (n = 41). Blood samples were collected at selected time points from 0 to 24 hours post injury/sham. Synovial fluid, intra-articular knee tissue and popliteal lymph nodes were collected at 24 hours. Cytokine/chemokine concentrations and gene expression were measured. Results Gene expression analysis revealed a rapid inflammatory response in the injured knees, accompanied by significantly increased levels of specific gene products in the synovial fluid; IL-1ß, TNFα, KC/GRO, IL-6, Eotaxin, MCP-1, MCP-3, MIP-1α, MIP-2, RANTES, A2M and AGP. Plasma analysis demonstrated significantly increased systemic levels of KC/GRO and IL-6 in injured rats already after 5 to 6 hours. Conclusion A rapid proinflammatory response, locally and systemically, characteristic of innate immunity, was demonstrated. Results reveal a more comprehensive inflammatory picture than previously shown, with resemblance to human haemophilic arthropathy, and with unique correlation between gene expression level, synovial concentration and plasma concentration in individual rats.


Asunto(s)
Citocinas/sangre , Hemartrosis/sangre , Hemofilia A/sangre , Mediadores de Inflamación/sangre , Inflamación/sangre , Articulaciones/metabolismo , Animales , Citocinas/genética , Modelos Animales de Enfermedad , Factor VIII/genética , Femenino , Predisposición Genética a la Enfermedad , Hemartrosis/etiología , Hemartrosis/genética , Hemofilia A/complicaciones , Hemofilia A/genética , Inflamación/etiología , Inflamación/genética , Ganglios Linfáticos/metabolismo , Masculino , Fenotipo , Ratas Transgénicas , Líquido Sinovial/metabolismo , Factores de Tiempo , Transcriptoma
11.
Blood ; 129(15): 2161-2171, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28039188

RESUMEN

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.


Asunto(s)
Factor IX , Hemartrosis , Hemofilia B , Articulaciones , Piel , Cicatrización de Heridas , Animales , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/farmacología , Hemartrosis/tratamiento farmacológico , Hemartrosis/genética , Hemartrosis/metabolismo , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemofilia B/metabolismo , Articulaciones/lesiones , Articulaciones/metabolismo , Ratones , Ratones Noqueados , Piel/lesiones , Piel/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
12.
Thromb Haemost ; 117(2): 277-285, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27929201

RESUMEN

Haemophilia A is a congenital bleeding disorder characterised by recurrent haemorrhages into the major joints. Haemophilic arthropathy is a well-established outcome of recurrent joint bleeding; however, it is clear that multiple factors determine the extent and severity of its occurrence. We sought to identify genetic factors related to abnormalities in range of motion (ROM) in the knees, ankles and elbows in a cohort of children and adolescents with haemophilia A not treated primarily with regular prophylaxis. Using data from the Haemophilia Growth and Development Study, we examined associations between 13,342 genetic markers and ROM scores measured at six-month intervals for up to seven years. As a first step, ordered logistic regression models were fit for each joint separately. A subset of SNP markers showing significant effects (p<0.01) on the right and left sides for at least two joints were included in a full model fit using a multivariate generalised linear mixed model assuming an ordinal response. The models contained all ROM scores obtained at all visits. Twenty-five markers analysed in the full model showed either increased or decreased risk of ROM abnormalities at the p<0.001 level. Several genes identified at either the first or second stage of the analysis have been associated with arthritis in a variety of large studies. Our results support the likelihood that risk for haemophilic arthropathy is associated with genetic factors, the identification of which holds promise for further advancing the individualisation of treatment.


Asunto(s)
Artritis/genética , Hemartrosis/genética , Hemofilia A/genética , Articulaciones/fisiopatología , Polimorfismo de Nucleótido Simple , Adolescente , Factores de Edad , Artritis/diagnóstico , Artritis/fisiopatología , Fenómenos Biomecánicos , Niño , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Hemartrosis/diagnóstico , Hemofilia A/complicaciones , Hemofilia A/diagnóstico , Humanos , Modelos Lineales , Modelos Logísticos , Estudios Longitudinales , Masculino , Análisis Multivariante , Oportunidad Relativa , Fenotipo , Pronóstico , Rango del Movimiento Articular , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Estados Unidos , Adulto Joven
13.
J Thromb Haemost ; 14(6): 1216-25, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27060449

RESUMEN

UNLABELLED: Essentials Validating the F8 rat as a new intermediate-size animal model of hemophilic arthropathy. Factor VIII (FVIII) treated F8(-/-) rats suffered induced hemarthrosis analyzed by histopathology. F8 (-/-) animals develop hemophilic arthropathy upon hemarthrosis, preventable by FVIII treatment. The F8 (-/-) rat presents as a new pharmacologic model of hemophilic arthropathy. SUMMARY: Background Translational animal models of hemophilia are valuable for determining the pathobiology of the disease and its co-morbidities (e.g. hemophilic arthropathy, HA). The biologic mechanisms behind the development of HA, a painful and debilitating condition, are not completely understood. We recently characterized a F8(-/-) rat, which could be a new preclinical model of HA. Objectives To establish the F8(-/-) rat as a model of HA by determining if the F8(-/-) rat develops HA resembling human HA after an induced joint bleed and whether a second joint bleed causes further disease progression. Methods Wild-type and F8(-/-) rats were treated with vehicle or recombinant human factor VIII (rhFVIII) prior to a needle-induced joint bleed. Joint swelling was measured prior to injury, the following 7 days and upon euthanasia. Histologic sections of the joint were stained, and athropathic changes identified and scored with regard to synovitis, bone remodelling, cartilage degradation and hemosiderin deposition. Results Vehicle-treated F8(-/-) rats experienced marked joint swelling and developed chronic degenerative joint changes (i.e. fibrosis of the subsynovial membrane, chondrocyte loss and excessive bone remodeling). Treatment with rhFVIII reduced or prevented swelling and degenerative joint changes, returning the F8(-/-) animals to a wild-type phenotype. Conclusion The hemophilic phenotype of the F8(-/-) rat resulted in a persistent hemarthrosis following an induced joint bleed. This caused development of HA resembling human HA, which was prevented by rhFVIII treatment, confirming the potential of the F8(-/-) rat as a model of HA.


Asunto(s)
Modelos Animales de Enfermedad , Factor VIII/genética , Hemartrosis/genética , Hemartrosis/patología , Animales , Remodelación Ósea , Cartílago/patología , Condrocitos/patología , Progresión de la Enfermedad , Factor VIII/administración & dosificación , Genotipo , Hemofilia A/genética , Hemorragia , Hemosiderina/química , Humanos , Artropatías , Fenotipo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Sinovitis/patología
14.
Haematologica ; 101(2): 219-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26494839

RESUMEN

Recent evidence suggests that patients with severe hemophilia B may have a less severe disease compared to severe hemophilia A. To investigate clinical, radiological, laboratory and histological differences in the arthropathy of severe hemophilia A and hemophilia B, 70 patients with hemophilia A and 35 with hemophilia B with at least one joint bleeding were consecutively enrolled. Joint bleedings (<10, 10-50, >50), regimen of treatment (prophylaxis/on demand), World Federation of Hemophilia, Pettersson and ultrasound scores, serum soluble RANK ligand and osteoprotegerin were assessed in all patients. RANK, RANK ligand and osteoprotegerin expression was evaluated in synovial tissue from 18 hemophilia A and 4 hemophilia B patients. The percentage of patients with either 10-50 or more than 50 hemarthrosis was greater in hemophilia A than in hemophilia B (P<0.001 and P=0.03, respectively), while that with less than 10 hemarthrosis was higher in hemophilia B (P<0.0001). World Federation of Hemophilia (36.6 vs. 20.2; P<0.0001) and ultrasound (10.9 vs. 4.3; P<0.0001) score mean values were significantly higher in hemophilia A patients. Serum osteoprotegerin and soluble RANK ligand were decreased in hemophilia A versus hemophilia B (P<0.0001 and P=0.006, respectively). Osteoprotegerin expression was markedly reduced in synovial tissue from hemophilia A patients. In conclusion, the reduced number of hemarthrosis, the lower World Federation of Hemophilia and ultrasound scores, and higher osteoprotegerin expression in serum and synovial tissue in hemophilia B suggest that hemophilia B is a less severe disease than hemophilia A. Osteoprotegerin reduction seems to play a pivotal role in the progression of arthropathy in hemophilia A.


Asunto(s)
Hemartrosis/patología , Hemofilia A/patología , Hemofilia B/patología , Osteoprotegerina/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Expresión Génica , Hemartrosis/complicaciones , Hemartrosis/diagnóstico por imagen , Hemartrosis/genética , Hemofilia A/complicaciones , Hemofilia A/diagnóstico por imagen , Hemofilia A/genética , Hemofilia B/complicaciones , Hemofilia B/diagnóstico por imagen , Hemofilia B/genética , Humanos , Cápsula Articular/química , Cápsula Articular/patología , Masculino , Persona de Mediana Edad , Osteoprotegerina/sangre , Ligando RANK/sangre , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/sangre , Receptor Activador del Factor Nuclear kappa-B/genética , Índice de Severidad de la Enfermedad , Ultrasonografía
15.
Am J Hematol ; 90(11): 1027-35, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26257191

RESUMEN

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.


Asunto(s)
Factor VIII/genética , Hemartrosis/patología , Hemofilia A/patología , Neovascularización Patológica/patología , Remodelación Vascular , Actinas/genética , Actinas/metabolismo , Animales , Tobillo/irrigación sanguínea , Tobillo/patología , Modelos Animales de Enfermedad , Articulación del Codo/irrigación sanguínea , Articulación del Codo/metabolismo , Articulación del Codo/patología , Endoglina , Factor VIII/metabolismo , Expresión Génica , Hemartrosis/genética , Hemartrosis/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Articulación de la Rodilla/irrigación sanguínea , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/patología , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Haemophilia ; 20(3): 426-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24354535

RESUMEN

Haemophilia A is associated with recurrent joint bleeding which leads to synovitis and debilitating arthropathy. Coagulation factor VIII level is an important determinant of bleed number and development of arthropathy . The aim of this study was to compare the haemophilia joint health score (HJHS) and Gilbert score with severity, age, thrombin generation (TG) and underlying mutation in a haemophilia A cohort which had minimal access to haemostatic replacement therapy. Ninety-two haemophilia A individuals were recruited from Pakistan. Age, age at first bleed, target joints, haemophilic arthropathy joints, HJHS and Gilbert score were recorded. A strong correlation was found between HJHS and Gilbert score (r = 0.98), both were significantly higher in severe (n = 59) compared with non-severe (n = 29) individuals before the age of 12 years (P ≤ 0.01) but not thereafter. When individuals were divided according to developmental age (<12 years, 12-16 years and >16 years), both HJHS and Gilbert score were significantly lower in the youngest group (P ≤ 0.001), there was no difference between 12-16 years and >16 years. In severe individuals there was no correlation between in vitro TG and joint score, whereas in non-severe individuals there was a weak negative correlation. In the severe group, no significant difference was observed for either joint score according to the underlying mutation type (inversion, missense, nonsense, frameshift). In this cohort of haemophilia A individuals with minimal access to haemostatic treatment, haemophilic arthropathy correlated with severity and age; among severe individuals, joint health scores did not relate to either the underlying mutation or in vitro TG.


Asunto(s)
Factor VIII/provisión & distribución , Hemartrosis/fisiopatología , Hemofilia A/fisiopatología , Trombina/biosíntesis , Adolescente , Adulto , Niño , Estudios de Cohortes , Factor VIII/administración & dosificación , Femenino , Hemartrosis/sangre , Hemartrosis/etiología , Hemartrosis/genética , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemofilia A/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Mutación , Pakistán , Adulto Joven
17.
J Thromb Haemost ; 11(2): 293-306, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23231432

RESUMEN

BACKGROUND: The present study was designed to investigate the molecular signaling events from onset of bleeding through the development of arthropathy in a murine model of hemophilia A. METHODS AND RESULTS: A sharp-injury model of hemarthrosis was used. A global gene expression array on joint-specific RNA isolated 3 h post-injury revealed nuclear factor-kappa B (NF-κB) as the major transcription factor triggering inflammation. As a number of genes encoding the cytokines, growth factors and hypoxia regulating factors are known to be activated by NF-κB and many of these are part of the pathogenesis of various joint diseases, we reasoned that NF-κB-associated pathways may play a crucial role in blood-induced joint damage. To further understand its role, we screened NF-κB-associated pathways between 1 h to 90 days after injury. After a single articular bleed, distinct members of the NF-κB family (NF-κB1/NF-κB2/RelA/RelB) and their responsive pro-inflammatory cytokines (IL-1ß/IL-6/IFNγ/TNFα) were significantly up-regulated (> 2 fold, P < 0.05) in injured vs. control joints at the various time-points analyzed (1 h/3 h/7 h/24 h). After multiple bleeds (days 30/60/75/90), there was increased expression of NF-κB-associated factors that contribute to hypoxia (HIF-1α, 3.3-6.5 fold), angiogenesis (VEGF-α, 2.5-4.4 fold) and chondrocyte damage (matrix metalloproteinase-13, 2.8-3.8 fold) in the injured joints. Micro RNAs (miR) that are known to regulate NF-κB activation (miRs-9 and 155), inflammation (miRs-16, 155 and 182) and apoptosis (miRs-19a, 155 and 186) were also differentially expressed (-4 to +13-fold) after joint bleeding, indicating that the small RNAs could modulate the arthropathy phenotype. CONCLUSIONS: These data suggest that NF-κB-associated signaling pathways are involved in the development of hemophilic arthropathy.


Asunto(s)
Hemartrosis/etiología , Hemofilia A/complicaciones , Articulación de la Rodilla/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factor VIII/genética , Factor VIII/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genotipo , Hemartrosis/sangre , Hemartrosis/genética , Hemartrosis/inmunología , Hemartrosis/patología , Hemofilia A/sangre , Hemofilia A/genética , Inflamación/sangre , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Articulación de la Rodilla/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , FN-kappa B/genética , Fenotipo , Reproducibilidad de los Resultados , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Factores de Tiempo
18.
Haemophilia ; 18(5): 818-23, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22533488

RESUMEN

Haemophilic arthropathy (HA) is one of the main complications of recurrent bleeding episodes in patients with severe haemophilia. However, the precise reasons making joints the predilected site of bleeding in patients with haemophilia are not fully understood. The objective of this project was to study the potential effect of synovium-derived thrombomodulin (TM) on the pathophysiology of haemarthroses. The concentration of TM and tissue factor pathway inhibitor (TFPI) was measured in knee synovial fluid of patients with haemophilia and controls. We used these concentrations of TM and TFPI in a thrombin generation (TG) model to analyse their in vitro effects on coagulation in plasma of six male controls and six severe haemophiliacs. The expression of TM in synovial tissue was also studied in controls and haemophiliacs. Patients with HA had significantly higher synovial fluid TFPI and TM levels, with a mean of 47 ± 27 ng/mL (P = 0.033) and 56 ± 25 ng/mL (P = 0.031), respectively, compared to the control group which presented lower levels of synovial fluid TFPI (26 ± 9 ng/mL) and TM concentrations (39 ± 21 ng/mL). TG capacity was significantly reduced in the presence of TM 56 ng/mL (P = 0.02), concentration observed in the synovial fluid of patients with HA. The concomitant addition of TM 56 ng/mL and TFPI 47 ng/mL induced a highly significant inhibition of TG in the same samples (P = 0.008).No significant inhibition of TG capacity was observed in the presence of control synovial concentration of TM (P > 0.05). Our results showed increased TM levels in synovial fluid and dramatically impaired expression of TM on synovial cells, suggesting a massive release of TM into the synovial fluid induced by a concerted action of neutrophils and cytokines on synovial cells as previously described in patients with rheumatoid arthritis.


Asunto(s)
Hemartrosis/etiología , Hemartrosis/fisiopatología , Hemofilia A/complicaciones , Hemofilia B/complicaciones , Trombomodulina/fisiología , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Células Cultivadas , Hemartrosis/genética , Hemofilia A/genética , Hemofilia A/fisiopatología , Hemofilia B/genética , Hemofilia B/fisiopatología , Humanos , Lipoproteínas/fisiología , Masculino , Estudios Prospectivos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Membrana Sinovial/patología , Membrana Sinovial/fisiopatología , Trombomodulina/genética
19.
J Thromb Haemost ; 8(2): 276-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19943872

RESUMEN

BACKGROUND: Large animal models that accurately mimic human hemophilia A (HA) are in great demand for developing and testing novel therapies to treat HA. OBJECTIVES: To re-establish a line of sheep exhibiting a spontaneous bleeding disorder closely mimicking severe human HA, fully characterize their clinical presentation, and define the molecular basis for disease. PATIENTS/METHODS: Sequential reproductive manipulations were performed with cryopreserved semen from a deceased affected ram. The resultant animals were examined for hematologic parameters, clinical symptoms, and responsiveness to human FVIII (hFVIII). The full coding region of sheep FVIII mRNA was sequenced to identify the genetic lesion. RESULTS AND CONCLUSIONS: The combined reproductive technologies yielded 36 carriers and 8 affected animals. The latter had almost non-existent levels of FVIII:C and extremely prolonged aPTT, with otherwise normal hematologic parameters. These animals exhibited bleeding from the umbilical cord, prolonged tail and nail cuticle bleeding time, and multiple episodes of severe spontaneous bleeding, including hemarthroses, muscle hematomas and hematuria, all of which responded to hFVIII. Inhibitors of hFVIII were detected in four treated animals, further establishing the preclinical value of this model. Sequencing identified a premature stop codon and frame-shift in exon 14, providing a molecular explanation for HA. Given the decades of experience using sheep to study both normal physiology and a wide array of diseases and the high homology between human and sheep FVIII, this new model will enable a better understanding of HA and facilitate the development and testing of novel treatments that can directly translate to HA patients.


Asunto(s)
Coagulación Sanguínea/genética , Factor VIII/genética , Hemartrosis/genética , Hemofilia A/genética , Factores de Edad , Envejecimiento , Animales , Secuencia de Bases , Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Codón sin Sentido , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exones , Factor VIII/metabolismo , Factor VIII/farmacología , Femenino , Predisposición Genética a la Enfermedad , Hemartrosis/sangre , Hemartrosis/tratamiento farmacológico , Hemartrosis/patología , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemofilia A/patología , Humanos , Masculino , Datos de Secuencia Molecular , Tiempo de Tromboplastina Parcial , Fenotipo , ARN Mensajero/sangre , Técnicas Reproductivas Asistidas , Ovinos , Especificidad de la Especie
20.
Haemophilia ; 15(4): 918-25, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19473418

RESUMEN

Type 3 von Willebrand disease (VWD) is a rare bleeding disorder with markedly decreased or absent von Willebrand factor (VWF) protein, accompanied by a parallel decrease in VWF function and factor VIII (FVIII) activity. The goal of this study was to describe the population of patients enrolled in the USA Centers for Disease Control Universal Data Collection (UDC) study with type 3 VWD, defined as a VWF:Ag of <10%, and to correlate bleeding symptoms with VWF and FVIII levels. Data on 150 patients were analysed. Almost all patients experienced bleeding episodes (98%) and required blood and/or factor product treatment (92%). While oral mucosal bleeding (the site of first bleed in 54%) was most common, subsequent muscle and joint bleeds were also seen (28%, 45%, respectively), and intracranial haemorrhage occurred in 8% of individuals. Mean age of first bleed was lower in those with either a FVIII < or =5% or a VWF:Ag <1%. Univariate marginal model analysis showed lower levels of FVIII and VWF:Ag both predicted a higher risk of joint bleeding. Longitudinal multivariate analysis found a lower FVIII level (P = 0.03), increasing age (P < 0.0001), history of joint bleeding (P = 0.001), higher body mass index (BMI) (P < 0.0001), and use of home infusion (P = 0.02) were all negatively associated with joint mobility. Low levels of VWF:Ag (P = 0.003) and male sex (P = 0.007) were also negatively associated with joint function. This study documents the strong bleeding phenotype in severe VWD and provides data to help target therapy, including prophylaxis, for patients most at risk of bleeding complications.


Asunto(s)
Hemartrosis/diagnóstico , Hemorragia/diagnóstico , Enfermedades de von Willebrand/diagnóstico , Adolescente , Niño , Preescolar , Técnicas de Laboratorio Clínico , Femenino , Genotipo , Hemartrosis/epidemiología , Hemartrosis/genética , Hemorragia/epidemiología , Hemorragia/genética , Humanos , Masculino , Índice de Severidad de la Enfermedad , Estados Unidos/epidemiología , Adulto Joven , Enfermedades de von Willebrand/epidemiología , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA