Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
J Dairy Sci ; 107(1): 9-23, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37678791

RESUMO

Milk fat globule epidermal growth factor 8 (MFG-E8) and whey protein have emerged as promising bionutrient supplements for enhancing skeletal muscle mass and function. In the present study, aging-related sarcopenia rat model was employed to elucidate the effects of the combined administration of MFG-E8 and whey protein on the catabolism and anabolism of gastrocnemius protein. Combined intervention led to notable enhancements in the antioxidative stress status and mitochondrial biogenesis capacity of gastrocnemius muscle fibers in the aging rats, concomitant with a significant inhibition of lipid accumulation. Moreover, the synergistic effect of MFG-E8 and whey protein was found to exert modulatory effects on key signaling pathways, including PI3K/Akt/PGC-1α pathway and MAPK/ERK signaling pathways in the gastrocnemius muscle of the aging rats. Specifically, this combined intervention was observed to promote mitochondrial biogenesis and regulate the expression of protein anabolism and catabolism-related regulators, thereby facilitating the alleviation of mitochondrial oxidative stress and enhancing biogenesis in gastrocnemius tissues. The findings of our study provide compelling evidence for the potential of MFG-E8 as a promising dietary supplement with antisarcopenic properties to ameliorate muscle protein metabolism disorders and mitigate mitochondrial-mediated myoblast apoptosis induced by oxidative stress.


Assuntos
Glicolipídeos , Glicoproteínas , Gotículas Lipídicas , Sarcopenia , Animais , Ratos , Fator VIII/farmacologia , Galactose/farmacologia , Proteínas do Leite/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sarcopenia/prevenção & controle , Sarcopenia/veterinária , Transdução de Sinais , Proteínas do Soro do Leite/farmacologia
2.
J Cancer Res Clin Oncol ; 149(19): 17467-17478, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37889308

RESUMO

PURPOSE: Bleomycin, etoposide, and cisplatin combination chemotherapy (BEP) improves the survival of patients with testicular cancer, but is associated with potentially life-threatening toxicities like pneumonitis and thromboembolic events. This study explored the effects of physical exercise in patients with testicular cancer during or after BEP-chemotherapy on pulmonary and vascular endothelial toxicity. METHODS: In this post hoc analysis of a multicenter randomized clinical trial (NCT01642680), patients with metastatic testicular cancer scheduled to receive BEP-chemotherapy were randomized to a 24-week exercise intervention, initiated during (group A) or after BEP-chemotherapy (group B). Endpoints were pulmonary function (forced vital capacity (FVC), forced expiratory volume in one second (FEV1), lung transfer-coefficient and transfer factor for carbon monoxide (KCO, DLCO) and markers of vascular endothelial dysfunction (von Willebrand factor (vWF) and factor VIII). RESULTS: Thirty patients were included. Post-chemotherapy, patients declined less in FVC, FEV1 and DLCO in group A compared to group B. Post-chemotherapy, vWF and factor VIII were significantly lower in group A compared to group B. After completion of exercise, started either during BEP-chemotherapy or thereafter, no between-group differences were found. At 1-year post-intervention, significant between-group differences were found in favour of group A in DLCO and KCO. CONCLUSIONS: Patients who exercised during BEP-chemotherapy better preserved FVC, FEV1 and DLCO, measured directly post-chemotherapy and 1-year post-intervention (DLCO, KCO). This coincided with less increase in vWF and factor VIII measured directly post-chemotherapy. These data support a beneficial role of a physical exercise intervention during BEP-chemotherapy on pulmonary and vascular damage in patients with testicular cancer. TRIAL REGISTRY: Optimal Timing of Physical Activity in Cancer Treatment (ACT) Registry URL: https://clinicaltrials.gov/ct2/show/NCT01642680 . TRIAL REGISTRATION NUMBER: NCT01642680.


Assuntos
Neoplasias Testiculares , Masculino , Humanos , Neoplasias Testiculares/tratamento farmacológico , Cisplatino , Etoposídeo , Bleomicina , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Fator de von Willebrand/farmacologia , Fator de von Willebrand/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Pulmão/patologia , Exercício Físico
3.
Exp Eye Res ; 234: 109572, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37451566

RESUMO

Our previous research shown that tumor necrosis factor-alpha-induced protein 8 (TNFAIP8) is elevated in the plasma extracellular vesicles and vitreous humor in diabetic retinopathy (DR). TNFAIP8 also significantly increases the viability of human retinal microvascular endothelial cells (HRMECs) and promotes cell migration and tube formation in vitro. To comprehensively explore its role in DR, we investigated the effect of TNFAIP8 on DR development using an animal model in this study. A TNFAIP8-overexpressing adeno-associated virus (AAV) vector and streptozotocin-induced mouse model was used. The AAV-TNFAIP8 vector was injected into the mice intravitreally, and the effect was evaluated. The evaluation included analysis of retinal structure and function using electroretinography, optical coherence tomography, and histological assessment. The influence of TNFAIP8 on the avascular area, retinal leukostasis, and the expression levels of inflammatory factors was also determined. TNFAIP8 significantly decreased a/b-wave amplitude and retinal thickness in diabetic mice. Histological assessment showed that TNFAIP8 aggravated pathological abnormalities with distorted organization of the retina. TNFAIP8 also significantly increased the avascular area, leukostasis, and the expression of inflammatory factors, such as TNFα, IL1ß, ICAM1, and GFAP, in the retina. The results of this study support the role of TNFAIP8 in DR pathogenesis. A mechanistic understanding of TNFAIP8 may offer novel therapeutic strategies.


Assuntos
Diabetes Mellitus Experimental , Retinopatia Diabética , Leucostasia , Camundongos , Humanos , Animais , Retinopatia Diabética/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Diabetes Mellitus Experimental/metabolismo , Fator VIII/metabolismo , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Células Endoteliais/metabolismo , Leucostasia/metabolismo , Retina/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo
4.
FASEB J ; 37(6): e22950, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37144883

RESUMO

Fracture nonunion and bone defects are challenging for orthopedic surgeons. Milk fat globule-epidermal growth factor 8 (MFG-E8), a glycoprotein possibly secreted by macrophages in a fracture hematoma, participates in bone development. However, the role of MFG-E8 in the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is unclear. We investigated the osteogenic effect of MFG-E8 in vitro and in vivo. The CCK-8 assay was used to assess the effect of recombinant human MFG-E8 (rhMFG-E8) on the viability of hBMSCs. Osteogenesis was investigated using RT-PCR, Western blotting, and immunofluorescence. Alkaline phosphatase (ALP) and Alizarin red staining were used to evaluate ALP activity and mineralization, respectively. An enzyme-linked immunosorbent assay was conducted to evaluate the secretory MFG-E8 concentration. Knockdown and overexpression of MFG-E8 in hBMSCs were established via siRNA and lentivirus vector transfection, respectively. Exogenous rhMFG-E8 was used to verify the in vivo therapeutic effect in a tibia bone defect model based on radiographic analysis and histological evaluation. Endogenous and secretory MFG-E8 levels increased significantly during the early osteogenic differentiation of hBMSCs. Knockdown of MFG-E8 inhibited the osteogenic differentiation of hBMSCs. Overexpression of MFG-E8 and rhMFG-E8 protein increased the expression of osteogenesis-related genes and proteins and enhanced calcium deposition. The active ß-catenin to total ß-catenin ratio and the p-GSK3ß protein level were increased by MFG-E8. The MFG-E8-induced enhanced osteogenic differentiation of hBMSCs was partially attenuated by a GSK3ß/ß-catenin signaling inhibitor. Recombinant MFG-E8 accelerated bone healing in a rat tibial-defect model. In conclusion, MFG-E8 promotes the osteogenic differentiation of hBMSCs by regulating the GSK3ß/ß-catenin signaling pathway and so, is a potential therapeutic target.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Ratos , Animais , Osteogênese/fisiologia , beta Catenina/genética , beta Catenina/metabolismo , Fator VIII/metabolismo , Fator VIII/farmacologia , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Transdução de Sinais/fisiologia , Diferenciação Celular/fisiologia , Glicoproteínas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas , Via de Sinalização Wnt , Células da Medula Óssea/metabolismo
5.
Blood Coagul Fibrinolysis ; 32(2): 87-102, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33186132

RESUMO

Cryoprecipitate (cryo) is a plasma-derived blood product utilized during trauma resuscitation, surgery, and other major bleeding. Although local quality control metrics exist, inherent donor variability, and processing may confer differences in hemostatic effect between sources. The purposes of this study were to quantify procoagulant content in three global sources of cryo and evaluate their functional hemostatic effect. In this Institutional Review Board exempt study, 24 units of group A cryo from three different sources, American Red Cross single donor and pooled donor, Australian Red Cross single donor, Southwestern United States single donor, and Southwest pooled donor, were evaluated. Procoagulant factors were quantified from each source using ELISA and automated clot-based assays. Functional hemostasis was evaluated using rotational Thromboelastometry (ROTEM). Microparticles isolated from cryo units were enumerated and evaluated for cellular origin by flow cytometry, as well as their capacity to support thrombin generation. Southwestern United States single donor units demonstrated highest levels of fibrinogen, fibronectin, factor VIII, and von Willebrand factor in the selected units. In the coagulopathy model, successive doses from all cryo units were significantly correlated to decreasing coagulation time (P = 0.0100), and increasing maximum clot firmness (P = 0.0002) and alpha angle (P = 0.0009). Southwest pooled donor demonstrated significantly shorter coagulation time at all three doses (P = 0.02) than other sources. Microparticles support prothrombinase activity and thrombin generation. In this study of global cryo sources, procoagulant activity and in-vitro clot formation varied by source. This could be explained by variance in production and storage protocols. Further study is warranted to assess functional variance in cryo to optimize and standardize the use of cryo products.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Coagulantes/química , Coagulantes/farmacologia , Fator VIII/química , Fator VIII/farmacologia , Fibrinogênio/química , Fibrinogênio/farmacologia , Fatores de Coagulação Sanguínea/análise , Fatores de Coagulação Sanguínea/farmacologia , Testes de Coagulação Sanguínea , Doadores de Sangue , Hemostasia/efeitos dos fármacos , Humanos
6.
Expert Rev Hematol ; 13(7): 731-743, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32573295

RESUMO

INTRODUCTION: Hemophilia A is a bleeding disorder traditionally managed with standard half-life (SHL) factor (F) VIII concentrates. Extended half-life (EHL) FVIII products and emicizumab-kywh, a nonfactor therapy, are newer treatment options. Additional nonfactor agents and gene therapy are expected to reach the market in the near future. AREAS COVERED: A PubMed (MEDLINE) search from 1962 to April 2020 related to hemophilia A, its management, and the products currently available for prophylaxis was performed to comprehensively review these topics and analyze the benefits and drawbacks of each therapeutic. EXPERT OPINION: Prophylaxis with SHL FVIII concentrates remains the standard of care for patients with severe hemophilia A and may also be considered for selected individuals with moderate disease. Several years of real-world experience with EHL FVIII, emicizumab-kywh, and other agents in development will be necessary to determine their ultimate roles in the prevention of bleeding and its complications. Gene therapy may not provide a permanent cure for hemophilia A.


Assuntos
Quimioprevenção , Hemofilia A/sangue , Hemofilia A/complicações , Hemorragia/etiologia , Hemorragia/prevenção & controle , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Quimioprevenção/métodos , Tomada de Decisão Clínica , Ensaios Clínicos como Assunto , Gerenciamento Clínico , Fator VIII/efeitos adversos , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Hemofilia A/diagnóstico , Hemofilia A/tratamento farmacológico , Hemorragia/diagnóstico , Humanos , Isoanticorpos/sangue , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Índice de Gravidade de Doença , Resultado do Tratamento
7.
Haemophilia ; 26(3): 478-486, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32338433

RESUMO

INTRODUCTION: Antihaemophilic factor (recombinant) (rAHF; ADVATE® ) is approved for prophylaxis and treatment of bleeding in children and adults with haemophilia A. Reconstitution in 2 mL sterile water for injection instead of 5 mL allows for a 60% reduction in infusion volume and administration time, but could increase the likelihood of hypersensitivity and infusion-related reactions, especially in children. AIM: To assess local tolerability, safety and effectiveness of rAHF 2 mL during routine clinical practice factor VIII (FVIII) replacement (on-demand and prophylaxis) in children with severe (FVIII < 1%) or moderately severe (FVIII 1%-2%) haemophilia A. METHODS: This was a prospective, non-interventional, postauthorization safety surveillance study (NCT02093741). Eligible patients were previously treated with rAHF and had a negative inhibitor test result during ≤10 exposure days prior to study entry. RESULTS: Of 65 patients enrolled (0-11 years of age), 54 and 11 had severe and moderately severe haemophilia A, respectively; 56 patients received prophylaxis, and 11 had ≤50 exposure days, of which 4 had ≤4 exposure days. No patients reported local hypersensitivity reactions, treatment-related adverse events or developed inhibitors. Investigators rated overall effectiveness of rAHF 2 mL prophylaxis as excellent or good. Ninety-four bleeding events in 34 patients were treated. Haemostatic effectiveness was rated as excellent or good for 75.8% of bleeds; 86.2% of bleeds required 1 or 2 infusions. CONCLUSION: In children with severe/moderately severe haemophilia A, no hypersensitivity reactions were reported with rAHF 2 mL treatment, and the safety and effectiveness are consistent with data previously reported for rAHF 5 mL.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Água/química , Criança , Pré-Escolar , Monitoramento Epidemiológico , Fator VIII/farmacologia , Feminino , Humanos , Lactente , Recém-Nascido , Injeções , Masculino
8.
Haemophilia ; 26(3): 450-458, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32293786

RESUMO

INTRODUCTION: Turoctocog alfa pegol is a glycoPEGylated recombinant factor VIII (FVIII) with an extended half-life developed for prophylaxis, treatment of bleeds and perioperative management in patients with haemophilia A. AIM: Evaluate the efficacy and safety of turoctocog alfa pegol treatment for major and minor surgeries in the pathfinder 3 and 5 phase III trials. METHODS: Adults/adolescents aged ≥12 years with severe haemophilia A (FVIII <1%) received perioperative turoctocog alfa pegol treatment planned to achieve FVIII activity levels >80% during major surgery (pathfinder 3). The primary end point was haemostatic efficacy during surgery; secondary end points were blood loss, haemostatic effect postsurgery, consumption, transfusions, safety and health economics. Children (0-11 years) undergoing minor surgeries received 20-75 IU/kg turoctocog alfa pegol at Investigator's discretion (pathfinder 5). RESULTS: pathfinder 3 included 35 patients undergoing 49 major surgeries. Haemostasis was successful in 47/49 (95.9%) surgeries; two had moderate haemostatic responses. Median (mean) blood loss during major surgery was 75 (322.6) mL. Four bleeds were reported postsurgery; three were successfully treated with turoctocog alfa pegol (one was not evaluated). On the day of surgery, overall mean (median) dose was 75.5 (74.5) IU/kg and mean (median) number of doses was 1.7 (2.0). Five procedures required 11 transfusions on the day of surgery or days 1-6. No safety concerns or inhibitors were identified. Forty-five minor surgeries in 23 children were performed without complications. CONCLUSION: Turoctocog alfa pegol was effective for perioperative haemostatic management of major and minor surgeries in patients across age groups with severe haemophilia A.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/cirurgia , Procedimentos Cirúrgicos Menores/métodos , Adolescente , Adulto , Idoso , Fator VIII/farmacologia , Feminino , Hemofilia A/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Adulto Jovem
9.
Haemophilia ; 25(6): 996-1002, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31584736

RESUMO

INTRODUCTION: The most severe side effect in haemophilia A treatment is the development of antifactor VIII antibodies, also called inhibitors. Why inhibitors develop in a proportion of treated patients while others are unaffected still remains unanswered. The presence of immunological danger signals, associated with events such as infection or surgery, has been proposed to play a role. Previous studies demonstrated that the presence of the bacterial molecule lipopolysaccharide (LPS) can synergistically increase the activation of human DC and subsequent T cell activation by FVIII. AIM AND METHODS: In the present study, we investigated whether a combination of two danger signals can further increase immune cell activation by FVIII. For this, human in vitro differentiated DC that were treated with combinations of danger signals were co-cultured with autologous primary T cells, and T cell proliferation was analysed. RESULTS: Interestingly, by combining LPS with a second danger signal, lower LPS concentrations were sufficient to synergistically increase DC and subsequent T cell activation by FVIII. Of note, a combination of LPS and the double-stranded RNA, polyinosinic-polycytidylic acid (poly(I:C)), was most potent in increasing FVIII immunogenicity, followed by LPS + R848 (resiquimod). However, a combination of LPS and the bacterial lipopeptide Pam3CysSK4 did not induce increased immune cell activation by FVIII. CONCLUSION: Thus, individual combinations of danger signals can increase FVIII product immunogenicity. This should be considered in the treatment routine of haemophilia A patients.


Assuntos
Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Humanos , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
10.
Thromb Res ; 183: 13-19, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31513978

RESUMO

INTRODUCTION: Ensuring hemostasis during invasive procedures is a challenge in patients with severe hemophilia A. This analysis evaluated efficacy and safety of BAY 94-9027, an extended-half-life recombinant factor VIII (FVIII), in the surgical setting. MATERIALS AND METHODS: Patients participating in an open-label BAY 94-9027 clinical trial who underwent major surgery were included in the analysis. Investigator/surgeon assessment of hemostasis during surgery was the primary outcome. In addition, information about FVIII use, FVIII levels during perioperative period, bleeding complications and FVIII inhibitor development were collected. RESULTS: Data were analyzed for 26 major surgeries (orthopedic, n = 21) in 20 patients aged 13-61 years. BAY 94-9027 provided effective hemostasis during all procedures. FVIII levels 6-8 h post preoperative infusion and prior to the first follow-up infusion were in the range expected to maintain protection in the major surgery setting. The median time from preoperative infusion to the first follow-up infusion (the first infusion administered after the preoperative infusion) was 12.33 (3.6-49.9) h. No intraoperative bleeding complications occurred, and no new inhibitors developed following any surgery. CONCLUSIONS: The results of the study demonstrate that BAY 94-9027 was efficacious and well tolerated in the treatment of patients undergoing major surgeries. Advantages of BAY 94-9027 include the potential for less frequent infusion and reduced factor consumption, which should simplify the management of patients during major surgery.


Assuntos
Coagulantes/uso terapêutico , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Polietilenoglicóis/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Adolescente , Adulto , Idoso , Criança , Coagulantes/farmacologia , Fator VIII/farmacologia , Feminino , Hemofilia A/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Polietilenoglicóis/farmacologia , Proteínas Recombinantes/farmacologia , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA