Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Agric Environ Med ; 31(2): 160-169, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38940098

RESUMEN

INTRODUCTION AND OBJECTIVE: As globalization and modernization continue to impact people's lives, a significant shift in lifestyle has taken place, resulting in a worldwide decrease in physical activity and an increase in unhealthy eating patterns. Physical inactivity has become the fourth leading cause of death globally. The aim of this scoping review is to analyze the concept and development of integrating physical activity into healthcare (IPAHc), based on the principles of sports and exercise medicine (SEM) and exercise is medicine (EIM). REVIEW METHODS: A systematic search was conducted of relevant published studies with full text using PubMed, Scopus, Web of Science, Academic Search Ultimate, Medline, and SPORTDiscus, via the EBSCO search platform. BRIEF DESCRIPTION OF THE STATE OF KNOWLEDGE: Twenty-nine studies met the inclusion criteria. The integration pathway centres around physical activity consultation and/or referral, and information technology which has been extensively utilized in IPAHc, including websites, electronic medical records, social media, wearable devices, mobile software, and referral tools. SEM and EIM face numerous implementation challenges, such as time constraints, education/training, resources, and tools. SUMMARY: The concept of IPAHc involves the integration of Physical Activity Vital Signs (PAVS) into electronic medical records to evaluate the physical activity levels of the general population. This can assist individuals in achieving fitness goals, preventing diseases, treating existing illnesses, and undergoing rehabilitation. IPAHc has been in development for many years and is now being explored in practice. Despite the widespread use of information technology in this integration process, a number of challenges still need addressing.


Asunto(s)
Ejercicio Físico , Humanos , Atención a la Salud
2.
Int J Pharm ; 659: 124291, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38821434

RESUMEN

Hemophilic arthropathy (HA) is a condition caused by recurrent intra-articular bleeding in patients with hemophilia. Pro-inflammatory cytokines play a crucial role in the pathogenesis of HA. Our previous research demonstrated that a novel compound, piperazino-enaminone (JODI), effectively inhibited pro-inflammatory cytokines, including IL-6, MCP-1, MIP-1α, and MIP-1ß, in a mouse model of hemarthrosis. This study aims to enhance the anti-inflammatory effect of JODI by employing nanoparticle delivery systems, which could potentially improve its poor water solubility. Here, we developed liposomes modified with polyethylene glycol (PEG) for the delivery of JODI (JODI-LIP), and found that JODI-LIP exhibited uniform size, morphology, good stability and in vitro release degree. JODI-LIP mitigated cytotoxicity of JODI, and significantly suppressed the production of pro-inflammatory cytokines (TNF-α and IL-1ß) and nitric oxide (NO) release in RAW 264.7 cells stimulated by lipopolysaccharide (LPS), as well as the proliferation of human fibroblast-like synovial (HFLS) cells. In a murine model of HA, JODI-LIP demonstrated superior efficacy in ameliorating joint swelling and synovitis, compared to JODI. Importantly, JODI-LIP markedly reduced pro-inflammatory cytokines (TNF-α, IFN-γ, IL-33, and MCP-1) in injured joints. No hepatic or hematological toxicity was observed in mice treated with JODI-LIP. In summary, our results suggest that JODI-LIP holds promise as a therapeutic intervention for HA by attenuating pro-inflammatory cytokine levels.


Asunto(s)
Antiinflamatorios , Citocinas , Modelos Animales de Enfermedad , Liposomas , Óxido Nítrico , Animales , Ratones , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Citocinas/metabolismo , Células RAW 264.7 , Humanos , Masculino , Óxido Nítrico/metabolismo , Hemartrosis/tratamiento farmacológico , Hemofilia A/tratamiento farmacológico , Piperazinas/farmacología , Piperazinas/administración & dosificación , Piperazinas/química , Polietilenglicoles/química , Polietilenglicoles/administración & dosificación , Lipopolisacáridos
3.
Front Public Health ; 11: 1232625, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38054068

RESUMEN

Physical activity (PA) is safe for most pregnant women, improving both maternal fitness and birth outcomes. Despite evidence of benefits, most pregnant women eliminate or reduce PA during pregnancy. This systematic review aimed to analyze the factors affecting maternal PA during pregnancy with reference to a socio-ecological model. A systematic search of relevant published studies between 2001 and 2022 was conducted through PubMed, Scopus, Web of Science, Academic Search Ultimate, Medline, and SPORTDiscus with full text via the EBSCO platform. A total of 32 studies that met the inclusion criteria were reviewed. The findings revealed that various study designs can lead to different outcomes in terms of what is identified as a PA facilitator or barrier. The factors that positively influenced PA in pregnant women were: higher levels of education, knowledge, and skills, as well as access to mass media. Conversely, lower levels of education, lack of knowledge and skills, low income, pregnancy discomforts, limited time, safety concerns, and societal perceptions of PA in pregnancy acted as barriers. Additionally, family, colleagues/friends, and partners could either support or hinder PA. Factors affecting overall maternal PA were somewhat different from those affecting the moderate-to-vigorous intensity of PA. Pregnant women receive little organizational and policy support. There is an urgent need to provide accessible information and resource systems for pregnant women. Since most pregnant women are motivated to engage in PA and susceptible to family advice, interventions should not be limited only to pregnant women, but should involve a family member, especially partners. There is a need to take global, systemic actions to promote an active lifestyle in pregnancy. Addressing safety concerns related to PA during pregnancy should be a significant part of these promotional activities.


Asunto(s)
Ejercicio Físico , Mujeres Embarazadas , Embarazo , Femenino , Humanos , Familia , Estilo de Vida
4.
Front Immunol ; 14: 1116084, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936967

RESUMEN

Objective: Rheumatoid arthritis (RA) is the most common form of autoimmune inflammatory arthritis. Intra-articular gene delivery to block proinflammatory cytokines has been studied in pre-clinical models and human clinical trials. It has been demonstrated that the level of programmed death-ligand 1 (PD-L1) is associated with rheumatoid arthritis (RA). This study examined the therapeutic role of PD-L1 by intra-articular delivery via adeno-associated virus (AAV) vectors in the mouse collagen-induced arthritis (CIA) model. Methods: Mice were intra-articularly injected with AAV5 vectors encoding human PD-L1 on day 0 and immunized with bovine type II collagen to induce CIA simultaneously. On day 49 post AAV administration, joints were collected for histo-pathological and cytokine analysis. Additionally, the systemic impacts of intra-articular injection of AAV5/PD-L1 vectors were also studied. To study the therapeutic effect of PD-L1, AAV5/PD-L1 vectors were administered into the joints of RA mice on day 21. Results: After administration of AAV5/PD-L1 vectors, strong PD-L1 expression was detected in AAV transduced joints. Joints treated with PD-L1 at the time of arthritis induction exhibited significantly less swelling and improved histopathological scores when compared to untreated joints. Additionally, the infiltration of T cells and macrophages was decreased in joints of CIA mice that received AAV5/PD-L1 vectors (P<0.05). The levels of pro-inflammatory cytokines, including IL-1, IL-6, IL-17 and TNFα, were lower in AAV5/PD-L1 treated than untreated joints (P<0.05). Furthermore, the administration of AAV5/PD-L1 vectors into the joints of CIA mice did not impact serum cytokine levels and the antibody titers to type II collagen. Biodistribution of AAV vectors after intra-articular injection showed undetectable AAV genomes in other tissues except for a low level in the liver. Similar to the results of AAV5/PD-L1 vector administration on day 0, decreased joint swelling and lower histopathological damage were observed in joints treated with AAV5/PD-L1 vectors on day 21. Conclusion: The results from this study demonstrate that local AAV mediated PD-L1 gene delivery into the joints is able to prevent the development and block the progression of arthritis in CIA mice without impacting systemic immune responses. This study provides a novel strategy to effectively treat inflammatory joint diseases using local AAV gene therapy by interference with immune checkpoint pathways.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Humanos , Ratones , Artritis Reumatoide/terapia , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Colágeno Tipo II/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/terapia , Distribución Tisular
5.
Nat Commun ; 13(1): 7275, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36434000

RESUMEN

AAV-delivered CRISPR/Cas9 (AAV-CRISPR) has shown promising potentials in preclinical models to efficiently insert therapeutic gene sequences in somatic tissues. However, the AAV input doses required were prohibitively high and posed serious risk of toxicity. Here, we performed AAV-CRISPR mediated homology-independent knock-in at a new target site in mAlb 3'UTR and demonstrated that single dose of AAVs enabled long-term integration and expression of hF9 transgene in both adult and neonatal hemophilia B mice (mF9 -/-), yielding high levels of circulating human Factor IX (hFIX) and stable hemostasis restoration during entire 48-week observation period. Furthermore, we achieved hemostasis correction with a significantly lower AAV dose (2 × 109 vg/neonate and 1 × 1010 vg/adult mouse) through liver-specific gene knock-in using hyperactive hF9R338L variant. The plasma antibodies against Cas9 and AAV in the neonatal mice receiving low-dose AAV-CRISPR were negligible, which lent support to the development of AAV-CRISPR mediated somatic knock-in for treating inherited diseases.


Asunto(s)
Hemofilia B , Ratones , Animales , Humanos , Hemofilia B/genética , Hemofilia B/terapia , Edición Génica , Sistemas CRISPR-Cas/genética , Formación de Anticuerpos , Vectores Genéticos/genética , Hemostasis , Hígado
6.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(5): 1549-1556, 2022 Oct.
Artículo en Chino | MEDLINE | ID: mdl-36208264

RESUMEN

OBJECTIVE: To explore the effect of lenalidomide on human fibroblast-like synovial cells (HFLS) and the therapeutic efficacy on hemophilic arthropathy in hemophilia A mice model. METHODS: In vitro, to remodel the inflammatory environment of synovial tissue after hemorrhage, ferric citrate and recombinant TNF-α were added into the cell culture medium of HFLS. Cell Counting Kit-8 (CCK-8), Enzyme-linked immunosorbent assay (ELISA), Quantitative Real-time PCR (RT-qPCR) and flow cytometry were employed for detection of the effects of lenalidomide on the proliferation ability, pro-inflammatory cytokines release and apoptosis of HFLS cells. In vivo, hemophilia arthropathy was remodeled in hemophilia A mice by induction of hemarthrosis. A series of doses of lenalidomide (0.1, 0.3 and 1.0 g/kg) was administrated intra-articularly. Tissues of knee joints were collected on the 14th day after administration, and the protective effect of lenalidomide on arthritis in hemophilia A mice were evaluated by RT-qPCR and histological grading. RESULTS: In vitro, compared with the untreated control group, lenalidomide could significantly inhibit the proliferation of HFLS cells (P<0.05), and the effect was the most significant when the concentration was 0.01 µmol/L (P<0.001). Compared with the control group, lenalidomide could significantly inhibit the expression levels of TNF-α, IL-1ß, IL-6 and IFN-γ in HFLS cells (P<0.05). The flow cytometry results showed that lenalidomide could enhance the apoptotis of HFLS cells (P<0.05). The results of RT-qPCR showed that lenalidomide could significantly reduce the mRNA expression levels of TNF-α, IL-1ß, IL-6,MCP-1 and VEGF in the joint tissues (P<0.05). Histological results showed that compared with the injured group, lenalidomide could significantly reduce the pathological sequela after hemarthrosis induction, e.g. synovial thickening and neo-angiogenesis in the synovium. The protection displayed a dose-response pattern roughly. CONCLUSION: In vitro, lenalidomide can inhibit the proliferation of HFLS cells, promote their apoptosis, and inhibit the expression of pro-inflammatory cytokines. In vivo, lenalidomide can significantly decrease the expression of pro-inflammatory cytokines in the joints of mice, and prevent the development of inflammation and neo-angiogenesis. The results provide a theoretical and experimental basis for the clinical application of lenalidomide in the treatment of hemophilic arthropathy.


Asunto(s)
Artritis , Hemofilia A , Animales , Citocinas/metabolismo , Hemartrosis/patología , Hemofilia A/genética , Humanos , Interleucina-6 , Lenalidomida , Ratones , Neovascularización Patológica , ARN Mensajero , Factor de Necrosis Tumoral alfa , Factor A de Crecimiento Endotelial Vascular
7.
Front Med (Lausanne) ; 9: 880763, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35991645

RESUMEN

Adeno-associated virus (AAV) gene therapy has been successfully applied in hemophilia patients excluding patients with inhibitors. During the coagulation pathway, activated factor V (FVa) functions downstream as a cofactor of activated factor X (FXa) to amplify thrombin generation. We hypothesize that the expression of FVa via gene therapy can improve hemostasis of both factor IX and FVIII deficiencies, regardless of clotting factor inhibitor. A human FVa (hFVa) expression cassette was constructed, and AAV8 vectors encoding hFVa (AAV8/TTR-hFVa) were intravenously administrated into mice with hemophilia A and B with or without FVIII inhibitors. Hemostasis, including hFVa level, activated partial thromboplastin time (aPTT), tail clip, and the saphenous vein bleeding assay (SVBA), was evaluated. In hemophilia B mice, a dose of 4 × 1013 vg/kg AAV8/TTR-hFVa vectors achieved a complete phenotypic correction over 28 weeks. In hemophilia A mice, hemostasis improvement was also achieved, regardless of FVIII inhibitor development. In vivo hemostasis efficacy was confirmed by tail clip and SVBA. Interestingly, while minimal shortening of aPTT was observed at a lower dose of AAV8 vectors, hemostasis improvement was still achieved via in vivo bleeding assays. Collectively, FVa-based AAV gene therapy shows promise for hemostasis correction in hemophilia, regardless of inhibitor development and no potential risk for thrombosis.

8.
Lancet Haematol ; 9(7): e504-e513, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35598604

RESUMEN

BACKGROUND: A novel, engineered, liver-tropic adeno-associated virus vector expressing a hyperactive Padua factor IX (FIX) protein (BBM-H901) has been developed and is promising for haemophilia B gene therapy. We aimed to explore its safety and activity in increasing FIX concentrations and reducing bleeding frequency. METHODS: We did a single-centre, single-arm, phase 1, pilot trial evaluating the safety and activity of a single intravenous infusion of BBM-H901 at the Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (Tianjin, China). We enrolled adult patients with haemophilia B (aged >18 years) with baseline FIX coagulation activity (FIX:C) of less than 2 IU/dL, no FIX inhibitor, and low titre of neutralising antibodies (≤1:4) against vector capsid. Eligible participants were intravenously infused with a single dose of 5 × 1012 vector genomes (vg)/kg of BBM-H901 after 1 week of prophylactic prednisone treatment (1 mg/kg per day). Primary endpoints were the incidence of treatment-related adverse events, change in alanine aminotransferase (ALT) and aspartate amino transferase (AST), and development of antibodies against vector capsid within 1 year of infusion. We report the results of the prespecified 1-year analysis following complete enrolment. The trial is registered with ClinicalTrials.gov, NCT04135300, and is complete. FINDINGS: Between Oct 16, 2019, and Jan 13, 2021, 12 male participants were assessed, and ten Chinese participants were enrolled and infused with BBM-H901. After a median follow-up of 58 weeks (IQR 51·5-99·5), mean FIX:C reached mean 36·9 IU/dL (SD 20·5). No serious adverse events, no grade 3-4 adverse events were observed. Grade 1-2 adverse events related to BBM-H901 include pyrexia (1 [10%]) and elevation of aminotransferase(1 [10%]). No FIX inhibitors were observed. All participants developed antibodies against vector capsid after infusion. Eight (80%) participants had ALT and AST concentrations below the upper limit of normal throughout the follow-up period. Two (20%) participants had elevation of ALT and AST accompanied with decrease of FIX:C, which remained at 7 IU/dL and 11.8 IU/dL, respectively. INTERPRETATION: This pilot study suggests that liver-tropic BBM-H901 is safe 1 year after infusion. Vector derived FIX:C concentration is sufficiently high to prevent bleeding events and minimise the need for replacement therapy in small populations with haemophilia B. These findings support further study. FUNDING: Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences, National Key Research and Development Program of China, National Natural Science Foundation of China, Tianjin Municipal Science and Technology Commission Grant, and Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences.


Asunto(s)
Factor IX , Hemofilia B , Adulto , Dependovirus/genética , Dependovirus/metabolismo , Factor IX/efectos adversos , Glucocorticoides/efectos adversos , Hemofilia B/tratamiento farmacológico , Hemorragia/inducido químicamente , Humanos , Hígado , Masculino , Proyectos Piloto
9.
Front Pharmacol ; 13: 815317, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35173619

RESUMEN

Adeno-associated virus (AAV) mediated gene therapy has been successfully applied in clinical trials, including hemophilia. Novel AAV vectors have been developed with enhanced transduction and specific tissue tropism. Considering the difference in efficacy of AAV transduction between animal models and patients, the chimeric xenograft mouse model with human hepatocytes has unique advantages of studying AAV transduction efficiency in human hepatocytes. However, it is unclear whether the results in humanized mice can predict AAV transduction efficiency in human hepatocytes. To address this issue, we studied the AAV transduction efficacy in canine hepatocytes in both canine hepatocyte xenografted mice and real dogs. After administration of AAV vectors from different serotypes into canine hepatocyte xenograft mice, AAV8 induced the best canine hepatocyte transduction followed by AAV9, then AAV3, 7, 5 and 2. After administration of AAV/cFIX (cFIX-opt-R338L) vectors in hemophilia B dogs, consistent with the result in chimeric mice, AAV8 induced the highest cFIX protein expression and function, followed by AAV9 and then AAV2. These results suggest that mice xenografted with hepatocytes from different species could be used to predict the AAV liver transduction in real species and highlight this potential platform to explore novel AAV variants for future clinical applications.

10.
Hum Gene Ther ; 33(5-6): 290-300, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34486389

RESUMEN

Neutralizing antibodies (NAbs) strongly limit adeno-associated virus (AAV) vector transduction and repeated administration. Previous studies have shown that NAbs induced by AAVs are associated with T and B cell activation and that the B7/CD28 and CD40/CD40L costimulation signaling pathways are involved. Cytotoxic T lymphocyte-associated antigen 4 (CTLA4) and CD40 are vital molecules that participate in the costimulatory pathway. In this study, we evaluated CTLA4-Ig and CD40-Ig immunosuppreve efficacies through AAV and investigated their effects on the feasibility for multiple systemic administrations of AAV vectors. The results showed that a single administration of AAV vector carrying either CTLA4-Ig alone or with CD40-Ig could greatly reduce the level of NAbs. An AAV serotype-specific immune tolerance could be successfully established, which enabled repeated, that is, second and third, systemic administration of AAV vectors in the same mice. A combination of CTLA4-Ig and CD40-Ig delivered via AAV vectors significantly inhibited T and B cell activations without affecting the immune response to the total immunoglobulin G production and cytokines. Interestingly, exogenous gene expression significantly improved after multiple administrations of AAV vector in vivo. Our study generates a reliable and effective method for repeated dosing of AAV vectors that is needed on gene therapy.


Asunto(s)
Dependovirus , Inmunoconjugados , Abatacept , Animales , Antígenos CD40/genética , Antígenos CD40/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Inmunoconjugados/genética , Ratones , Linfocitos T/metabolismo
11.
Blood Coagul Fibrinolysis ; 32(8): 584-590, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34693916

RESUMEN

Subclinical bleeding is a haemorrhage event not clinically detected in haemophilia, and no reliable method is available for predicting subclinical bleeding. We investigated whether haemophilia mice have subclinical haemorrhage and evaluated potential biomarkers including multiple cytokine changes to predict subclinical haemorrhage. Plasma from naïve FVIII-/- and FIX-/- mice and their wild-type counterparts (FVIII WT and FIX WT, respectively) were measured for prothrombin fragment 1 + 2 (F1 + 2) and multiple cytokines. Haemophilia mice with induced hemarthrosis were used as positive clinical bleeding controls. Naive haemophilia mice that displayed higher levels than positive bleeding control were counted. Univariate and multivariate analyses of cytokines were performed. Compared with wild-type mice (FVIII WT 1.1-6.2 vs. FIX WT 2.7-6.7 pmol/l), F1 + 2 widely varied in both haemophilia mouse strains (FVIII-/- 3.7-25.7 vs. FIX-/- 2.7-15.7 pmol/l). Each cytokine varied widely in both naive haemophilia A and B mice, but not significantly, for most cytokines. In comparison to haemophilia mice with hemarthrosis bleeding challenge, naive FVIII-/- mice had elevated pro-inflammatory cytokines and FIX-/- mice had elevated anti-inflammatory cytokines. In addition, interleukin (IL)-4, followed by IL-1, IL-6, TNF-α and MIP-1α in FVIII-/- mice and MIP-1α, followed by IL-1, IL-10 in FVIII-/- mice exhibited significant differences potentially associated with potential subclinical bleeding. Naive haemophilia mice showed elevated pro-inflammatory cytokines with different patterns, represented by pro-inflammatory cytokine elevation in more naïve FVIII-/- mice and more anti-inflammatory cytokines in FIX-/- mice.


Asunto(s)
Hemofilia A , Animales , Citocinas , Factor VIII/genética , Hemartrosis , Hemofilia A/genética , Hemorragia , Ratones
12.
Hum Gene Ther ; 31(7-8): 448-458, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32079420

RESUMEN

Hemophilia arthropathy (HA) represents the majority of morbidity in severe hemophilia patients, especially in resource-limited countries. Adeno-associated virus (AAV)-mediated gene therapy is showing promise for managing hemophilia. However, patients with neutralizing antibodies (NAbs) against AAV, and inhibitors to clotting factors, are excluded from such therapy. This study explored the feasibility of AAV-mediated local gene therapy for HA. Factor VIII knockout (FVIII-/-) mice, with or without a FVIII inhibitor, were subjected to hemarthrosis induction and treated with either intravenous (IV) or intraarticular (IA) recombinant human factor VIII (rhFVIII). To investigate whether rhFVIII carried the risk to develop a FVIII inhibitor, FVIII-/- mice were treated with three doses of IV or IA rhFVIII and inhibitor development was measured. In patients with established HA requiring synovial fluid aspiration, plasma, and synovial fluid were collected and measured for anti-AAV capsid IgG (serotypes 1-9 and 843) and NAbs for AAV843. IA rhFVIII provided better protection from synovitis compared with IV rhFVIII, with or without the FVIII inhibitor. While IV rhFVIII led to all FVIII-/- mice developing an FVIII inhibitor (n = 31, median 4.9 Bethesda units [BU]/mL), only 50% of the mice developed a FVIII inhibitor by IA administration, and at a lower titer (median 0.55 BU/mL). In hemophilia patients, total anti-AAV IgG was lowest for AAV4 and AAV5, both in plasma and synovial fluid. Anti-AAV IgGs in synovial fluid for most samples were lower or similar to the plasma levels. These results show that direct IA rhFVIII administration yields better protection against bleeding-induced joint damage, even in the presence of an inhibitor antibody. IA rhFVIII delivery carried a lower risk of FVIII inhibitor formation compared with IV FVIII. The anti-AAV antibody level in synovial fluid was similar or lower than the plasma level, supporting the feasibility of local gene therapy for managing HA.


Asunto(s)
Dependovirus , Factor VIII/genética , Factor VIII/uso terapéutico , Terapia Genética/métodos , Hemartrosis/terapia , Hemofilia A/terapia , Animales , Estudios de Factibilidad , Humanos , Ratones , Ratones Noqueados , Proteínas Recombinantes/uso terapéutico , Sinovitis/terapia
13.
Sci Rep ; 9(1): 14428, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594977

RESUMEN

While joint damage is the primary co-morbidity of hemophilia, osteoporosis and osteopenia are also observed. Coagulation factor VIII deficient (FVIII-/-) mice develop an osteoporotic phenotype in the absence of induced hemarthrosis that is exacerbated two weeks after an induced joint injury. Here we have compared comprehensively the bone health of clotting factor VIII, factor IX, and Von Willebrand Factor knockout (FVIII-/-, FIX-/-, and VWF-/- respectively) mice both in the absence of joint hemorrhage and following induced joint injury. We found FVIII-/- and FIX-/- mice, but not VWF-/- mice, developmentally have an osteoporotic phenotype. Unilateral induced hemarthrosis causes further bone damage in both FVIII-/- and FIX-/- mice, but has little effect on VWF-/- bone health, indicating that the FVIII.VWF complex is not required for normal bone remodeling in vivo. To further investigate the bone healing following hemarthrosis in hemophilia we examined a two week time course using microCT, serum chemistry, and histological analysis. Elevated ratio of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL), increased osterix+ osteoblastic cells, and decreased smoothness of the cortical bone surface were evident within several days of injury, indicative of acute heterotopic mineralization along the cortical surface. This was closely followed by increased interleukin-6 (IL-6) levels, increased osteoclast numbers, and significant trabecular bone loss. Uncoupled and disorganized bone formation and resorption continued for the duration of the study resulting in significant deterioration of the joint. Further elucidation of the shared mechanisms underlying abnormal bone homeostasis in the absence of FVIII or FIX is needed to guide evidence-based approaches to the screening and treatment of the prevalent bone defects in hemophilia A and B.


Asunto(s)
Factor IX/genética , Factor VIII/genética , Hemofilia A/metabolismo , Hemofilia B/metabolismo , Factor de von Willebrand/genética , Animales , Pruebas de Coagulación Sanguínea , Huesos/metabolismo , Hemofilia A/genética , Hemofilia A/patología , Hemofilia B/genética , Hemofilia B/patología , Humanos , Interleucina-6/genética , Masculino , Ratones , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/patología , Osteoporosis/genética , Osteoporosis/patología , Fenotipo , Ligando RANK/genética , Factor de Transcripción Sp7/genética
14.
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
15.
Inflammation ; 42(5): 1719-1729, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31129747

RESUMEN

Hemarthrosis is the primary cause of hemophiliac arthropathy (HA). Pro-inflammatory cytokines are thought to play an important role in the pathogenesis of HA, and thus, anti-cytokine approaches may be used as an adjuvant therapy. A novel series of enaminone compounds (JODI), that contain the N-aryl piperazino motif, have been shown in vitro to reduce pro-inflammatory cytokines and thus may be efficacious in vivo. In this report, we will assess whether JODI can suppress multiple cytokines which might be potentially responsible for joint inflammation in a mouse model of hemarthrosis. The results showed that JODI significantly improved the survival after LPS treatment, and most pro-inflammatory cytokines/chemokines were decreased significantly after JODI administration. In the hemophilia mouse model, hemarthrosis resulted in local cytokine/chemokine changes, represented by elevated pro-inflammatory (IL-6, MCP-1, MIP-1α, MIP-1ß) and pro-angiogenic (VEGF and IL-33) cytokines, and decreased anti-pro-inflammatory cytokines IL-4 and IL-10. The changes were reversed by administration of JODI, which can be used as a novel approach to manage hemophilia arthropathy.


Asunto(s)
Citocinas/efectos de los fármacos , Hemartrosis/tratamiento farmacológico , Hemofilia A/complicaciones , Cetonas/química , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hemartrosis/etiología , Hemartrosis/patología , Inflamación/prevención & control , Cetonas/farmacología , Cetonas/uso terapéutico , Ratones , Neovascularización Patológica/prevención & control , Piperazina/química , Piperazina/farmacología , Piperazina/uso terapéutico
16.
Int J Hematol ; 110(1): 59-68, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31006077

RESUMEN

Bleeding into the joints represents the major morbidity of severe hemophilia and predisposes it to hemophilic arthropathy (HA). In a reproducible hemarthrosis mouse model, we found distinct changes in thrombin activity in joint tissue homogenate following exposure of the joint to blood in wide type (WT) and hemophilic B mice. Specifically, at early time points (4 h and 24 h) after hemarthrosis, thrombin activity in WT mice quickly peaked at 4 h, and returned to baseline after 1 week. In hemophilia B mice, there was no/minimal thrombin activity in joint tissues at 4 h and 24 h, whereas at 72 h and thereafter, thrombin activity kept rising, and persisted at a higher level. Nevertheless, prothrombin had not decreased in both WT and hemophilia. The pattern was also confirmed by Western blotting and immunostaining. To optimize the protection against development of HA, we tested different treatment regimens by administration of clotting factor IX into hemophilia B mouse after hemarthrosis induction, including a total of 600 IU/kg FIX within the first 24 h or the whole 2-week period. We concluded that timely (in the first 24 h) and sufficient hemostasis correction is critical for a better protection against the development of hemophilic arthropathy.


Asunto(s)
Factor IX/uso terapéutico , Hemartrosis/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Hemofilia B/complicaciones , Hemofilia B/patología , Hemorragia/tratamiento farmacológico , Articulaciones/patología , Ratones , Trombina/metabolismo , Factores de Tiempo
17.
J Clin Med ; 9(1)2019 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-31892201

RESUMEN

Repeated intra-articular hemorrhages lead to hemophilic arthropathy in severe hemophilia. Inflammation and pro-inflammatory cytokines (e.g., tumor necrosis factor alpha (TNFα)) might be involved in this pathogenesis. We hypothesized that anti-TNFα may provide adjuvant protection for hemophilic arthropathy management. We measured TNFα in synovial lavage from hemophilia mice subjected to hemarthrosis induction and synovial fluid from patients with hemophilic arthropathy (n = 5). In hemophilia mice, recurrent hemarthroses were induced, anti-TNFα was initiated either from day (D)7 after one hemarthrosis episode or D21 after three hemarthroses episodes (n ≥ 7/treatment group). In patients with hemophilic arthropathy (16 patients with 17 affected joints), a single dose of anti-TNFα was administered intra-articularly. Efficacy, characterized by synovial membrane thickness and vascularity, was determined. Elevated TNFα in synovial lavage was found in the hemophilia mice and patients with hemophilic arthropathy. Hemophilia mice subjected to three hemarthroses developed severe synovitis (Synovitis score of 6.0 ± 1.6). Factor IX (FIX) replacement alone partially improved the pathological changes (Synovitis score of 4.2 ± 0.8). However, anti-TNFα treatment initiated at D7, not D21, significantly provided protection (Synovitis score of 1.8 ± 0.9 vs. 3.9 ± 0.3). In patients with hemophilic arthropathy, intra-articular anti-TNFα significantly decreased synovial thickness and vascularity during the observed period from D7 to D30. Collectively, this preliminary study seems to indicate that TNFα may be associated with the pathogenicity of hemophilic arthropathy and anti-TNFα could provide adjuvant protection against hemophilic arthropathy. Further studies are required to confirm the preliminary results shown in this study.

18.
Mol Ther Methods Clin Dev ; 10: 257-267, 2018 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-30140713

RESUMEN

Adeno-associated virus (AAV) vectors have been successfully applied in hemophilia clinical trials. However, this approach is limited to patients without AAV-neutralizing antibodies (NAbs). In this study, we explored the feasibility of AAV re-administration in hemophilia A dogs treated initially 8 years ago with AAV8.canine FVIII. After the re-administration in two NAb-negative dogs with AAV8 vectors carrying human factor VIII (hFVIII), along with the proteasome inhibitor bortezomib, we observed a phenotypic improvement in both dogs that persisted in one dog. Phenotypic improvement disappeared at 59 days after re-administration in the other dog, and specific cytotoxic T lymphocytes (CTLs) to the capsid were detected at day 17, but not to hFVIII. hFVIII inhibitors were observed at day 59 and gradually increased. Mechanistic studies demonstrated an increase in pro-inflammatory cytokines, a decrease in immunomodulatory cytokines, as well as lower Tregs after re-administration. These results suggest that hFVIII inhibitor development may contribute to the therapeutic failure via immune response activation. Interestingly, it takes about 30-50 days for AAV NAb titers to decrease by half. Collectively, this study suggests that re-administration of the same AAV serotype after long-term follow-up is feasible and that the study of AAV NAb kinetics will provide important information for predicating the efficacy of re-administration.

19.
JCI Insight ; 3(12)2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29925692

RESUMEN

Data from clinical trials for hemophilia B using adeno-associated virus (AAV) vectors have demonstrated decreased transgenic coagulation factor IX (hFIX) expression 6-10 weeks after administration of a high vector dose. While it is likely that capsid-specific cytotoxic T lymphocytes eliminate vector-transduced hepatocytes, thereby resulting in decreased hFIX, this observation is not intuitively consistent with restored hFIX levels following prednisone application. Although the innate immune response is immediately activated following AAV vector infection via TLR pathways, no studies exist regarding the role of the innate immune response at later time points after AAV vector transduction. Herein, activation of the innate immune response in cell lines, primary human hepatocytes, and hepatocytes in a human chimeric mouse model was observed at later time points following AAV vector transduction. Mechanistic analysis demonstrated that the double-stranded RNA (dsRNA) sensor MDA5 was necessary for innate immune response activation and that transient knockdown of MDA5, or MAVS, decreased IFN-ß expression while increasing transgene production in AAV-transduced cells. These results both highlight the role of the dsRNA-triggered innate immune response in therapeutic transgene expression at later time points following AAV transduction and facilitate the execution of effective strategies to block the dsRNA innate immune response in future clinical trials.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/inmunología , Inmunidad Innata/inmunología , Infecciones por Parvoviridae/inmunología , ARN Bicatenario/inmunología , Transducción Genética , Animales , Cápside , Línea Celular , Factor IX/genética , Factor IX/metabolismo , Técnicas de Silenciamiento del Gen , Terapia Genética , Vectores Genéticos/genética , Células HEK293 , Células HeLa , Células Hep G2 , Hepatocitos/inmunología , Humanos , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón beta/metabolismo , Hígado/metabolismo , Ratones , Modelos Animales , ARN Bicatenario/genética , Transgenes/genética , Trasplante Heterólogo
20.
Hum Gene Ther ; 29(3): 381-389, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28922951

RESUMEN

The development of inhibitory autoantibodies to the infused clotting factor VIII (FVIII) is a major complication for severe hemophilia A management. Novel therapy options for hemophilia have significantly progressed in the last decade, and a gene therapy cure for hemophilia is becoming a reality. However, mechanistic studies of FVIII autoantibodies (FVIII inhibitors) have lagged behind and remain a challenge for both protein replacement and gene therapy. FVIII inhibitor formation is assumed to be a classical T cell-dependent immune response in which cytokines/chemokines play an important role. The study of cytokine profile changes during FVIII inhibitor development may be helpful to understand the mechanism of inhibitor development and to explore potential novel approaches that will minimize the risk. After FVIII-/- mice were treated with intravenous administration of an adeno-associated virus 8 vector encoding human FVIII, FVIII expression peaked at week 2 (W2), and FVIII inhibitor was thoroughly developed at week 8 (W8). W8 plasma that showed positive FVIII inhibitor, and W2 samples with negative FVIII inhibitor (anti-FVIII[+]), were subjected to multiplex cytokines measurement. W8 and W2 samples were both negative for FVIII inhibitor (anti-FVIII[-]) as the control. In comparison to mice in the anti-FVIII(-) group, mice in the anti-FVIII(+) group exhibited significantly elevated pro-inflammatory cytokines of interleukin (IL)-1, IL-6, IL-12p40, monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1, MIP-2, and tumor necrosis factor alpha (TNF-α), especially at higher titers. The anti-inflammatory cytokine of transforming growth factor beta (TGF-ß) was decreased at W2 in both groups. Multivariate analysis of the risk factors for FVIII inhibitor development showed peak FVIII activity at W2. IL-6 and TNF-α at W8 were positively correlated with inhibitor formation, and negatively correlated with the age starting gene therapy. Collectively, the elevated monocyte derived pro-inflammatory cytokines/chemokines, together with the decreased anti-inflammatory cytokine of TGF-ß at an early time point, may contribute to the persistent inflammatory environment in favor of an immune response toward FVIII inhibitor development.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/inmunología , Citocinas/inmunología , Dependovirus , Factor VIII , Terapia Genética , Hemofilia A , Animales , Modelos Animales de Enfermedad , Factor VIII/genética , Factor VIII/inmunología , Hemofilia A/genética , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos , Ratones , Ratones Noqueados , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA