Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Gene Ther ; 30(3-4): 245-254, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-33456057

RESUMEN

Von Willebrand disease (VWD), the most common inherited bleeding disorder in humans, is caused by quantitative or qualitative defects in von Willebrand factor (VWF). VWD represents a potential target for gene therapy applications, as a single treatment could potentially result in a long-term correction of the disease. In recent years, several liver-directed gene therapy approaches have been exploited for VWD, but their efficacy was generally limited by the large size of the VWF transgene and the reduced hemostatic activity of the protein produced from hepatocytes. In this context, we aimed at developing a gene therapy strategy for gene delivery into endothelial cells, the natural site of biosynthesis of VWF. We optimized an endothelial-specific dual hybrid AAV vector, in which the large VWF cDNA was put under the control of an endothelial promoter and correctly reconstituted upon cell transduction by a combination of trans-splicing and homologous recombination mechanisms. In addition, we modified the AAV vector capsid by introducing an endothelial-targeting peptide to improve the efficiency for endothelial-directed gene transfer. This vector platform allowed the reconstitution of full-length VWF transgene both in vitro in human umbilical vein endothelial cells and in vivo in VWD mice, resulting in long-term expression of VWF.


Asunto(s)
Enfermedades de von Willebrand , Factor de von Willebrand , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Enfermedades de von Willebrand/genética , Enfermedades de von Willebrand/metabolismo , Enfermedades de von Willebrand/terapia , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Vectores Genéticos
2.
Arterioscler Thromb Vasc Biol ; 41(1): e1-e17, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232198

RESUMEN

OBJECTIVE: TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, such as, platelet adhesion to vascular endothelium, nitric oxide/cGMP (cyclic guanosine monophosphate) signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1-/- mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. CONCLUSIONS: Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.


Asunto(s)
Arteriopatías Oclusivas/prevención & control , Antígeno CD47/antagonistas & inhibidores , Fibrinolíticos/farmacología , Péptidos Cíclicos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Trombosis/prevención & control , Trombospondina 1/antagonistas & inhibidores , Animales , Arteriopatías Oclusivas/sangre , Arteriopatías Oclusivas/metabolismo , Antígeno CD47/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/toxicidad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos Cíclicos/toxicidad , Inhibidores de Agregación Plaquetaria/toxicidad , Ratas Sprague-Dawley , Transducción de Señal , Trombosis/sangre , Trombosis/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factores de Tiempo
3.
Thromb Haemost ; 121(3): 261-269, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32987444

RESUMEN

Hemostasis is a tightly regulated process characterized by a finely tuned balance between procoagulant and anticoagulant systems. Among inherited hemostatic conditions, hemophilia is one of the most well-known bleeding disorders. Hemophilia A (HA) and B (HB) are due to deficiencies in coagulation factor VIII (FVIII) or FIX, respectively, leading to unwanted bleeding. Until recently, hemophilia treatment has consisted of prophylactic replacement therapy using plasma-derived or recombinant FVIII in cases of HA or FIX in cases of HB. Because FVIII and FIX deficiencies lead to an imbalance between procoagulant and anticoagulant systems, a recent upcoming strategy implies blocking of endogenous anticoagulant proteins to compensate for the procoagulant factor deficit, thus restoring hemostatic equilibrium. Important physiological proteins of the anticoagulant pathways belong to the serpin (serine protease inhibitor) family and, recently, different experimental and clinical studies have demonstrated that targeting natural serpins could decrease bleeding in hemophilia. Here, we aim to review the different, recent studies demonstrating that blocking serpins such as antithrombin, protein Z-dependent protease inhibitor, and protease nexin-1 or modifying a serpin like α1-antitrypsin could rebalance coagulation in hemophilia. Furthermore, we underline the potential therapeutic use of serpins for the treatment of hemophilia.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Serpinas/metabolismo , Serpinas/uso terapéutico , Animales , Descubrimiento de Drogas , Hemofilia A/sangre , Hemofilia A/metabolismo , Hemofilia B/sangre , Hemofilia B/metabolismo , Humanos , Serpinas/sangre
4.
Arterioscler Thromb Vasc Biol ; 40(12): 2860-2874, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32967458

RESUMEN

OBJECTIVE: Obesity is characterized by chronic low-grade inflammation and consequentially a hypercoagulable state, associating with an increased incidence of venous thromboembolism. Increased VWF (von Willebrand factor) plasma concentration and procoagulant function are independent risk factors for venous thromboembolism and are elevated in obese patients. Here, we explore the pathobiological role of VWF in obesity-associated venous thrombosis using murine models. Approach and Results: We first showed that diet-induced obese mice have increased VWF plasma levels and FVIII (factor VIII) activity compared with littermate controls. Elevated VWF levels appeared to be because of both increased synthesis and impaired clearance. Diet-induced obesity-associated venous thrombosis was assessed using the inferior vena cava-stenosis model of deep vein thrombosis. Diet-induced obese mice developed larger venous thrombi that were rich in VWF, erythrocytes, and leukocytes. Administering a polyclonal anti-VWF antibody or an anti-VWF A1 domain nanobody was protective against obesity-mediated thrombogenicity. Delayed administration (3 hours post-inferior vena cava stenosis) similarly reduced thrombus weight in diet-induced obese mice. CONCLUSIONS: This study demonstrates the critical role of VWF in the complex, thrombo-inflammatory state of obesity. It adds to the growing rationale for targeting VWF-specific interactions in thrombotic disease.


Asunto(s)
Dieta Alta en Grasa , Obesidad/complicaciones , Vena Cava Inferior/metabolismo , Trombosis de la Vena/etiología , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/genética , Proteína ADAMTS13/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Fibrinolíticos/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Transducción de Señal , Anticuerpos de Dominio Único/farmacología , Vena Cava Inferior/efectos de los fármacos , Vena Cava Inferior/patología , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología , Trombosis de la Vena/prevención & control , Factor de von Willebrand/antagonistas & inhibidores , Factor de von Willebrand/genética
5.
Thromb Haemost ; 120(9): 1300-1312, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32726852

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) is associated with a high cardiovascular mortality due to increased rates of vascular lesions and thrombotic events, as well as serum accumulation of uremic toxins. A subgroup of these toxins (advanced glycation end products [AGEs] and S100 proteins) can interact with the receptor for AGEs (RAGE). In this study, we analyzed the impact of CKD on platelet function and arterial thrombosis, and the potential role of RAGE in this process. METHODS: Twelve weeks after induction of CKD in mice, platelet function and time to complete carotid artery occlusion were analyzed in four groups of animals (sham-operated, CKD, apolipoprotein E [Apoe]-/-, and Apoe-/-/Ager-/- mice). RESULTS: Analysis of platelet function from whole blood and platelet-rich plasma showed hyperactivation of platelets only in CKD Apoe-/- mice. There was no difference when experiments were done on washed platelets. However, preincubation of such platelets with AGEs or S100 proteins induced RAGE-mediated platelet hyperactivation. In vivo, CKD significantly reduced carotid occlusion times of Apoe-/- mice (9.2 ± 1.1 vs. 11.1 ± 0.6 minutes for sham, p < 0.01). In contrast, CKD had no effect on occlusion times in Apoe-/-/Ager-/- mice. Moreover, carotid occlusion in Apoe-/- CKD mice occurred significantly faster than in Apoe-/-/Ager-/- CKD mice (p < 0.0001). CONCLUSION: Our results show that CKD induces platelet hyperactivation, accelerates thrombus formation in a murine model of arterial thrombosis, and that RAGE deletion has a protective role. We propose that RAGE ligands binding to RAGE is involved in CKD-induced arterial thrombosis.


Asunto(s)
Plaquetas/patología , Activación Plaquetaria , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Insuficiencia Renal Crónica/complicaciones , Trombosis/complicaciones , Animales , Plaquetas/metabolismo , Eliminación de Gen , Ratones Endogámicos C57BL , Receptor para Productos Finales de Glicación Avanzada/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
6.
J Thromb Haemost ; 18(9): 2155-2168, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32495984

RESUMEN

BACKGROUND: Protease nexin-1 (PN-1) is a member of the serine protease inhibitor (Serpin)-family, with thrombin as its main target. Current polyclonal and monoclonal antibodies against PN-1 frequently cross-react with plasminogen activator inhibitor-1 (PAI-1), a structurally and functionally homologous Serpin. OBJECTIVES: Here, we aimed to develop inhibitory single-domain antibodies (VHHs) that show specific binding to both human (hPN-1) and murine (mPN-1) PN-1. METHODS: PN-1-binding VHHs were isolated via phage-display using llama-derived or synthetic VHH-libraries. Following bacterial expression, purified VHHs were analyzed in binding and activity assays. RESULTS AND CONCLUSIONS: By using a llama-derived library, 2 PN-1 specific VHHs were obtained (KB-PN1-01 and KB-PN1-02). Despite their specificity, none displayed inhibitory activity toward hPN-1 or mPN-1. From the synthetic library, 4 VHHs (H12, B11, F06, A08) could be isolated that combined efficient binding to both hPN-1 and mPN-1 with negligible binding to PAI-1. Of these, B11, F06, and A08 were able to fully restore thrombin activity by blocking PN-1. As monovalent VHH, half-maximal inhibitory concentration values for hPN-1 were 50 ± 10, 290 ± 30, and 960 ± 390 nmol/L, for B11, F06, and A08, respectively, and 1580 ± 240, 560 ± 130, and 2880 ± 770 nmol/L for mPN-1. The inhibitory potential was improved 4- to 7-fold when bivalent VHHs were engineered. Importantly, all VHHs could block PN-1 activity in plasma as well as PN-1 released from activated platelets, one of the main sources of PN-1 during hemostasis. In conclusion, we report the generation of inhibitory anti-PN-1 antibodies using a specific approach to avoid cross-reactivity with the homologous Serpin PAI-1.


Asunto(s)
Anticuerpos de Dominio Único , Trombina , Animales , Anticuerpos Monoclonales , Técnicas de Visualización de Superficie Celular , Humanos , Ratones , Serpina E2/genética
7.
Blood ; 136(6): 740-748, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32369559

RESUMEN

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor IX/administración & dosificación , Factor X/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Modelos Animales , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Quimioterapia Combinada , Factor IX/análisis , Factor IX/inmunología , Factor VIII/administración & dosificación , Factor VIII/análisis , Factor VIII/uso terapéutico , Factor X/análisis , Factor X/inmunología , Factor XIa/farmacología , Femenino , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemofilia A/inmunología , Hemorragia/etiología , Infusiones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiempo de Tromboplastina Parcial , Cola (estructura animal)/lesiones , Trombina/biosíntesis
8.
EMBO Mol Med ; 12(4): e11298, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32159286

RESUMEN

Novel therapies for hemophilia, including non-factor replacement and in vivo gene therapy, are showing promising results in the clinic, including for patients having a history of inhibitor development. Here, we propose a novel therapeutic approach for hemophilia based on llama-derived single-domain antibody fragments (sdAbs) able to restore hemostasis by inhibiting the antithrombin (AT) anticoagulant pathway. We demonstrated that sdAbs engineered in multivalent conformations were able to block efficiently AT activity in vitro, restoring the thrombin generation potential in FVIII-deficient plasma. When delivered as a protein to hemophilia A mice, a selected bi-paratopic sdAb significantly reduced the blood loss in a model of acute bleeding injury. We then packaged this sdAb in a hepatotropic AAV8 vector and tested its safety and efficacy profile in hemophilic mouse models. We show that the long-term expression of the bi-paratopic sdAb in the liver is safe and poorly immunogenic, and results in sustained correction of the bleeding phenotype in hemophilia A and B mice, even in the presence of inhibitory antibodies to the therapeutic clotting factor.


Asunto(s)
Anticoagulantes , Antitrombinas , Hemofilia A , Anticuerpos de Dominio Único , Animales , Anticoagulantes/farmacología , Antitrombinas/farmacología , Coagulación Sanguínea/efectos de los fármacos , Hemofilia A/tratamiento farmacológico , Humanos , Ratones , Anticuerpos de Dominio Único/farmacología
9.
Blood ; 134(19): 1632-1644, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31383642

RESUMEN

Hemophilia A and B, diseases caused by the lack of factor VIII (FVIII) and factor IX (FIX) respectively, lead to insufficient thrombin production, and therefore to bleeding. New therapeutic strategies for hemophilia treatment that do not rely on clotting factor replacement, but imply the neutralization of natural anticoagulant proteins, have recently emerged. We propose an innovative approach consisting of targeting a natural and potent thrombin inhibitor, expressed by platelets, called protease nexin-1 (PN-1). By using the calibrated automated thrombin generation assay, we showed that a PN-1-neutralizing antibody could significantly shorten the thrombin burst in response to tissue factor in platelet-rich plasma (PRP) from patients with mild or moderate hemophilia. In contrast, in PRP from patients with severe hemophilia, PN-1 neutralization did not improve thrombin generation. However, after collagen-induced platelet activation, PN-1 deficiency in F8-/-mice or PN-1 blocking in patients with severe disease led to a significantly improved thrombin production in PRP, underlining the regulatory role of PN-1 released from platelet granules. In various bleeding models, F8-/-/PN-1-/- mice displayed significantly reduced blood loss and bleeding time compared with F8-/-mice. Moreover, platelet recruitment and fibrin(ogen) accumulation were significantly higher in F8-/-/PN-1-/- mice than in F8-/-mice in the ferric chloride-induced mesenteric vessel injury model. Thromboelastometry studies showed enhanced clot stability and lengthened clot lysis time in blood from F8-/-/PN-1-/- and from patients with hemophilia A incubated with a PN-1-neutralizing antibody compared with their respective controls. Our study thus provides proof of concept that PN-1 neutralization can be a novel approach for future clinical care in hemophilia.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/enzimología , Serpina E2/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Trastornos de la Coagulación Sanguínea Heredados/complicaciones , Hemorragia/etiología , Hemostasis/efectos de los fármacos , Humanos , Ratones , Ratones Noqueados , Activación Plaquetaria/efectos de los fármacos
10.
J Thromb Haemost ; 17(12): 2035-2046, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31423724

RESUMEN

BACKGROUND: Activated factor VII (FVIIa) is pertinent to the initiation of blood coagulation. Proteolytic and amidolytic activity of FVIIa are greatly enhanced by its cofactor, tissue factor (TF). OBJECTIVE: We aimed to generate a single-domain antibody (sdAb) that recognizes free FVIIa rather than TF-bound FVIIa. METHODS: A llama-derived phage library was used to screen for anti-FVIIa sdAbs. RESULTS: One sdAb, KB-FVIIa-004, bound to FVIIa, but not to its precursor FVII or to homologous proteins (prothrombin, factor X, or their activated derivatives). FVIIa amidolytic activity was inhibited by KB-FVIIa-004 (Ki  = 28-45 nM) in a competitive manner. KB-FVIIa-004 also inhibited FVIIa-mediated FX activation (Ki  = 26 nM). In contrast, KB-FVIIa-004 was inefficient in prolonging the clotting time of the prothrombin time-test, which was prolonged by a maximum of 10 s at high sdAb concentrations (10 µM). Furthermore, FVIIa/TF amidolytic activity or FVIIa/TF-mediated FX activation remained unaffected up to a 50-fold to 1000-fold molar excess of KB-FVIIa-004. These data suggest that KB-FVIIa-004 loses its inhibitory activity in the presence of TF. A KB-FVIIa-004/albumin fusion-protein (004-HSA) was generated for in vivo testing. By using 004-HSA, we observed that this sdAb blocked the therapeutic capacity of FVIIa to correct bleeding in FVIII-deficient mice. DISCUSSION: This observation is compatible with the view that FVIIa functions independently of TF under these conditions. In conclusion, we have generated a sdAb that specifically blocks TF-independent activity of FVIIa. This antibody can be used to gain insight into the roles of TF-bound and TF-free FVIIa.


Asunto(s)
Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Factor VIIa/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Tromboplastina/metabolismo , Animales , Coagulantes/antagonistas & inhibidores , Coagulantes/farmacología , Modelos Animales de Enfermedad , Factor VIII/genética , Factor VIIa/inmunología , Factor VIIa/metabolismo , Factor VIIa/farmacología , Femenino , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Humanos , Masculino , Ratones Endogámicos C57BL , Unión Proteica
11.
Haematologica ; 104(12): 2493-2500, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30819911

RESUMEN

Patients with type 2B von Willebrand disease (vWD) (caused by gain-of-function mutations in the gene coding for von Willebrand factor) display bleeding to a variable extent and, in some cases, thrombocytopenia. There are several underlying causes of thrombocytopenia in type 2B vWD. It was recently suggested that desialylation-mediated platelet clearance leads to thrombocytopenia in this disease. However, this hypothesis has not been tested in vivo The relationship between platelet desialylation and the platelet count was probed in 36 patients with type 2B von Willebrand disease (p.R1306Q, p.R1341Q, and p.V1316M mutations) and in a mouse model carrying the severe p.V1316M mutation (the 2B mouse). We observed abnormally high elevated levels of platelet desialylation in both patients with the p.V1316M mutation and the 2B mice. In vitro, we demonstrated that 2B p.V1316M/von Willebrand factor induced more desialylation of normal platelets than wild-type von Willebrand factor did. Furthermore, we found that N-glycans were desialylated and we identified αIIb and ß3 as desialylation targets. Treatment of 2B mice with sialidase inhibitors (which correct platelet desialylation) was not associated with the recovery of a normal platelet count. Lastly, we demonstrated that a critical platelet desialylation threshold (not achieved in either 2B patients or 2B mice) was required to induce thrombocytopenia in vivo In conclusion, in type 2B vWD, platelet desialylation has a minor role and is not sufficient to mediate thrombocytopenia.


Asunto(s)
Plaquetas/patología , Mutación , Ácido N-Acetilneuramínico/química , Trombocitopenia/patología , Enfermedad de von Willebrand Tipo 2/complicaciones , Factor de von Willebrand/genética , Animales , Plaquetas/metabolismo , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Integrina alfa2beta1/metabolismo , Integrina beta3/metabolismo , Masculino , Ratones , Ácido N-Acetilneuramínico/metabolismo , Recuento de Plaquetas , Polisacáridos/metabolismo , Pronóstico , Procesamiento Proteico-Postraduccional , Trombocitopenia/etiología , Trombocitopenia/metabolismo , Enfermedad de von Willebrand Tipo 2/genética , Enfermedad de von Willebrand Tipo 2/patología
12.
Cell Mol Life Sci ; 76(4): 791-807, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30498996

RESUMEN

In addition to its critical role in lysosomes for catabolism of sialoglycoconjugates, NEU1 is expressed at the plasma membrane and regulates a myriad of receptors by desialylation, playing a key role in many pathophysiological processes. Here, we developed a proteomic approach dedicated to the purification and identification by LC-MS/MS of plasma membrane NEU1 interaction partners in human macrophages. Already known interaction partners were identified as well as several new candidates such as the class B scavenger receptor CD36. Interaction between NEU1 and CD36 was confirmed by complementary approaches. We showed that elastin-derived peptides (EDP) desialylate CD36 and that this effect was blocked by the V14 peptide, which blocks the interaction between bioactive EDP and the elastin receptor complex (ERC). Importantly, EDP also increased the uptake of oxidized LDL by macrophages that is blocked by both the V14 peptide and the sialidase inhibitor 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA). These results demonstrate, for the first time, that binding of EDP to the ERC indirectly modulates CD36 sialylation level and regulates oxidized LDL uptake through this sialidase. These effects could contribute to the previously reported proatherogenic role of EDP and add a new dimension in the regulation of biological processes through NEU1.


Asunto(s)
Aterosclerosis , Antígenos CD36/metabolismo , Neuraminidasa/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Antígenos CD36/genética , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Elastina/química , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacología , Neuraminidasa/genética , Péptidos/metabolismo , Péptidos/farmacología , Unión Proteica , Proteómica/métodos , Interferencia de ARN , Células THP-1
13.
Blood ; 132(11): 1193-1197, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30064978

RESUMEN

Von Willebrand factor (VWF) modulates factor VIII (FVIII) clearance and the anti-FVIII immune response. Despite the high affinity that defines the FVIII/VWF interaction, association/dissociation kinetics dictates 2% to 5% FVIII being present as free protein. To avoid free FVIII when studying the FVIII-VWF complex in vivo, we designed a FVIII-nanobody fusion protein, with the nanobody part being directed against VWF. This fusion protein, designated FVIII-KB013bv, had a 25-fold higher affinity compared with B-domainless FVIII (BDD-FVIII) for VWF. In vitro analysis revealed full cofactor activity in 1-stage clotting and chromogenic assays (activity/antigen ratio 1.0 ± 0.3 and 1.1 ± 0.3, respectively). In vivo, FVIII-013bv displayed a twofold increased mean residence time compared with BDD-FVIII (3.0 hours vs 1.6 hours). In a tail clip-bleeding assay performed 24 hours after FVIII infusion, blood loss was significantly reduced in mice receiving FVIII-KB013bv vs BDD-FVIII (15 ± 7 µL vs 194 ± 146 µL; P = .0043). Unexpectedly, when examining anti-FVIII antibody formation in FVIII-deficient mice, the immune-response toward FVIII-KB013bv was significantly reduced compared with BDD-FVIII (1/8 vs 14/16 mice produced anti-FVIII antibodies after treatment with FVIII-KB013bv and BDD-FVIII, respectively). Our data show that a stabilized interaction between FVIII and VWF is associated with a prolonged survival of FVIII and a reduced immune response against FVIII.


Asunto(s)
Formación de Anticuerpos/efectos de los fármacos , Autoanticuerpos , Factor VIII , Proteínas Recombinantes de Fusión , Anticuerpos de Dominio Único/farmacología , Factor de von Willebrand , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Factor VIII/inmunología , Factor VIII/farmacocinética , Factor VIII/farmacología , Ratones , Ratones Mutantes , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Factor de von Willebrand/inmunología , Factor de von Willebrand/metabolismo
14.
Sci Rep ; 6: 38363, 2016 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-27917893

RESUMEN

Neuraminidase 1 (NEU1) is a lysosomal sialidase catalyzing the removal of terminal sialic acids from sialyloconjugates. A plasma membrane-bound NEU1 modulating a plethora of receptors by desialylation, has been consistently documented from the last ten years. Despite a growing interest of the scientific community to NEU1, its membrane organization is not understood and current structural and biochemical data cannot account for such membrane localization. By combining molecular biology and biochemical analyses with structural biophysics and computational approaches, we identified here two regions in human NEU1 - segments 139-159 (TM1) and 316-333 (TM2) - as potential transmembrane (TM) domains. In membrane mimicking environments, the corresponding peptides form stable α-helices and TM2 is suited for self-association. This was confirmed with full-size NEU1 by co-immunoprecipitations from membrane preparations and split-ubiquitin yeast two hybrids. The TM2 region was shown to be critical for dimerization since introduction of point mutations within TM2 leads to disruption of NEU1 dimerization and decrease of sialidase activity in membrane. In conclusion, these results bring new insights in the molecular organization of membrane-bound NEU1 and demonstrate, for the first time, the presence of two potential TM domains that may anchor NEU1 in the membrane, control its dimerization and sialidase activity.


Asunto(s)
Membrana Celular/química , Neuraminidasa/química , Fosfatidilcolinas/química , Ácidos Siálicos/química , Ubiquitina/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células COS , Membrana Celular/enzimología , Sistema Libre de Células/química , Sistema Libre de Células/metabolismo , Chlorocebus aethiops , Escherichia coli/química , Expresión Génica , Humanos , Modelos Moleculares , Neuraminidasa/genética , Neuraminidasa/metabolismo , Fosfatidilcolinas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ácidos Siálicos/metabolismo , Homología Estructural de Proteína , Especificidad por Sustrato , Ubiquitina/genética , Ubiquitina/metabolismo
15.
Front Pharmacol ; 7: 32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26973522

RESUMEN

Elastin, one of the longest-lived proteins, confers elasticity to tissues with high mechanical constraints. During aging or pathophysiological conditions such as cancer progression, this insoluble polymer of tropoelastin undergoes an important degradation leading to the release of bioactive elastin-derived peptides (EDPs), named elastokines. EDP exhibit several biological functions able to drive tumor development by regulating cell proliferation, invasion, survival, angiogenesis, and matrix metalloproteinase expression in various tumor and stromal cells. Although, several receptors have been suggested to bind elastokines (αvß3 and αvß5 integrins, galectin-3), their main receptor remains the elastin receptor complex (ERC). This heterotrimer comprises a peripheral subunit, named elastin binding protein (EBP), associated to the protective protein/cathepsin A (PPCA). The latter is bound to a membrane-associated protein called Neuraminidase-1 (Neu-1). The pro-tumoral effects of elastokines have been linked to their binding onto EBP. Additionally, Neu-1 sialidase activity is essential for their signal transduction. Consistently, EDP-EBP interaction and Neu-1 activity emerge as original anti-tumoral targets. Interestingly, besides its direct involvement in cancer progression, the ERC also regulates diabetes outcome and thrombosis, an important risk factor for cancer development and a vascular process highly increased in patients suffering from cancer. In this review, we will describe ERC and elastokines involvement in cancer development suggesting that this unique receptor would be a promising therapeutic target. We will also discuss the pharmacological concepts aiming at blocking its pro-tumoral activities. Finally, its emerging role in cancer-associated complications and pathologies such as diabetes and thrombotic events will be also considered.

16.
Cardiovasc Res ; 110(3): 298-308, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009176

RESUMEN

Cardiovascular diseases (CVDs) are the leading cause of death worldwide and represent a major problem of public health. Over the years, life expectancy has considerably increased throughout the world, and the prevalence of CVD is inevitably rising with the growing ageing of the population. The normal process of ageing is associated with progressive deterioration in structure and function of the vasculature, commonly called vascular ageing. At the vascular level, extracellular matrix (ECM) ageing leads to molecular alterations in long half-life proteins, such as elastin and collagen, and have critical effects on vascular diseases. This review highlights ECM alterations occurring during vascular ageing with a specific focus on elastin fragmentation and also the contribution of elastin-derived peptides (EDP) in age-related vascular complications. Moreover, current and new pharmacological strategies aiming at minimizing elastin degradation, EDP generation, and associated biological effects are discussed. These strategies may be of major relevance for preventing and/or delaying vascular ageing and its complications.


Asunto(s)
Envejecimiento/metabolismo , Arterias/metabolismo , Elastina/metabolismo , Matriz Extracelular/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedades Vasculares/metabolismo , Envejecimiento/patología , Animales , Arterias/efectos de los fármacos , Arterias/patología , Fármacos Cardiovasculares/uso terapéutico , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Humanos , Terapia Molecular Dirigida , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/metabolismo , Elastasa Pancreática/antagonistas & inhibidores , Elastasa Pancreática/metabolismo , Proteolisis , Inhibidores de Serina Proteinasa/uso terapéutico , Transducción de Señal/efectos de los fármacos , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/patología
17.
Arterioscler Thromb Vasc Biol ; 34(12): 2570-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25341794

RESUMEN

OBJECTIVE: Elastin is the major structural extracellular matrix component of the arterial wall that provides the elastic recoil properties and resilience essential for proper vascular function. Elastin-derived peptides (EDP) originating from elastin fragmentation during vascular remodeling have been shown to play an important role in cell physiology and development of cardiovascular diseases. However, their involvement in thrombosis has been unexplored to date. In this study, we investigated the effects of EDP on (1) platelet aggregation and related signaling and (2) thrombus formation. We also characterized the mechanism by which EDP regulate thrombosis. APPROACH AND RESULTS: We show that EDP, derived from organo-alkaline hydrolysate of bovine insoluble elastin (kappa-elastin), decrease human platelet aggregation in whole blood induced by weak and strong agonists, such as ADP, epinephrine, arachidonic acid, collagen, TRAP, and U46619. In a mouse whole blood perfusion assay over a collagen matrix, kappa-elastin and VGVAPG, the canonical peptide recognizing the elastin receptor complex, significantly decrease thrombus formation under arterial shear conditions. We confirmed these results in vivo by demonstrating that both kappa-elastin and VGVAPG significantly prolonged the time for complete arteriole occlusion in a mouse model of thrombosis and increased tail bleeding times. Finally, we demonstrate that the regulatory role of EDP on thrombosis relies on platelets that express a functional elastin receptor complex and on the ability of EDP to disrupt plasma von Willebrand factor interaction with collagen. CONCLUSIONS: These results highlight the complex nature of the mechanisms governing thrombus formation and reveal an unsuspected regulatory role for circulating EDP in thrombosis.


Asunto(s)
Elastina/fisiología , Trombosis/etiología , Animales , Plaquetas/fisiología , Catepsina A/sangre , Bovinos , Colágeno/sangre , Elastina/sangre , Elastina/química , Humanos , Ratones , Neuraminidasa/sangre , Oligopéptidos/sangre , Oligopéptidos/química , Oligopéptidos/fisiología , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Fragmentos de Péptidos/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteolisis , Receptores de Superficie Celular/sangre , Transducción de Señal , Trombosis/sangre , Remodelación Vascular/fisiología , Factor de von Willebrand/metabolismo
18.
Diabetes ; 62(11): 3807-16, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23919962

RESUMEN

Although it has long been established that the extracellular matrix acts as a mechanical support, its degradation products, which mainly accumulate during aging, have also been demonstrated to play an important role in cell physiology and the development of cardiovascular and metabolic diseases. In the current study, we show that elastin-derived peptides (EDPs) may be involved in the development of insulin resistance (IRES) in mice. In chow-fed mice, acute or chronic intravenous injections of EDPs induced hyperglycemic effects associated with glucose uptake reduction and IRES in skeletal muscle, liver, and adipose tissue. Based on in vivo, in vitro, and in silico approaches, we propose that this IRES is due to interaction between the insulin receptor (IR) and the neuraminidase-1 subunit of the elastin receptor complex triggered by EDPs. This interplay was correlated with decreased sialic acid levels on the ß-chain of the IR and reduction of IR signaling. In conclusion, this is the first study to demonstrate that EDPs, which mainly accumulate with aging, may be involved in the insidious development of IRES.


Asunto(s)
Elastina/metabolismo , Resistencia a la Insulina/fisiología , Fragmentos de Péptidos/farmacología , Animales , Metabolismo Energético/efectos de los fármacos , Hiperglucemia/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/análogos & derivados , Ácido N-Acetilneuramínico/farmacología , Neuraminidasa/metabolismo , Oligopéptidos/farmacología , Receptor de Insulina/metabolismo , Receptores de Superficie Celular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...