Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 22(5): 1489-1495, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38325597

RESUMEN

BACKGROUND: The recruitment of activated factor VIII (FVIII) at the surface of activated platelets is a key step toward the burst of thrombin and fibrin generation during thrombus formation at the site of vascular injury. It involves binding to phosphatidylserine and, possibly, to fibrin-bound αIIbß3. Seminal work had shown the binding of FVIII to resting platelets, yet without a clear understanding of a putative physiological relevance. OBJECTIVES: To characterize the effects of FVIII-platelet interaction and its potential modulation of platelet function. METHODS: FVIII was incubated with washed platelets. The effects on platelet activation (spontaneously or triggered by collagen and thrombin) were studied by flow cytometry and light transmission aggregometry. We explored the involvement of downstream pathways by studying phosphorylation profiles (Western blot). The FVIII-glycoprotein (GP) VI interaction was investigated by ELISA, confocal microscopy, and proximity ligation assay. RESULTS: FVIII bound to the surface of resting and activated platelets in a dose-dependent manner. FVIII at supraphysiological concentrations did not induce platelet activation but rather specifically inhibited collagen-induced platelet aggregation and altered glycoprotein VI (GPVI)-dependent phosphorylation. FVIII, freed of its chaperone protein von Willebrand factor (VWF), interacted in close proximity with GPVI at the platelet surface. CONCLUSION: We showed that VWF-free FVIII binding to, or close to, GPVI modulates platelet activation in vitro. This may represent an uncharacterized negative feedback loop to control overt platelet activation. Whether locally activated FVIII concentrations achieved during platelet accumulation and thrombus formation at the site of vascular injury in vivo are compatible with such a function remains to be determined.


Asunto(s)
Plaquetas , Factor VIII , Activación Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria , Humanos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Activación Plaquetaria/efectos de los fármacos , Plaquetas/metabolismo , Fosforilación , Factor VIII/metabolismo , Colágeno/metabolismo , Unión Proteica , Citometría de Flujo , Trombina/metabolismo , Relación Dosis-Respuesta a Droga , Microscopía Confocal
2.
J Thromb Haemost ; 21(10): 2776-2783, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37473843

RESUMEN

BACKGROUND: Emicizumab is a bispecific, chimeric, humanized immunoglobulin G (IgG)4 that mimics the procoagulant activity of factor (F) VIII (FVIII). Its long half-life and subcutaneous route of administration have been life-changing in treating patients with hemophilia A (HA) with or without FVIII inhibitors. However, emicizumab only partially mimics FVIII activity; it prevents but does not treat acute bleeds. Emergency management is particularly complicated in patients with FVIII inhibitors receiving emicizumab prophylaxis in whom exogenous FVIII is inefficient. We have shown recently that Imlifidase (IdeS), a bacterial IgG-degrading enzyme, efficiently eliminates human anti-FVIII IgG in a mouse model of severe HA with inhibitors and opens a therapeutic window for the administration of exogenous FVIII. OBJECTIVES: To investigate the impact of IdeS treatment in inhibitor-positive HA mice injected with emicizumab. METHODS: IdeS was injected to HA mice reconstituted with human neutralizing anti-FVIII IgG and treated with emicizumab. RESULTS: IdeS hydrolyzed emicizumab in vitro and in vivo, albeit, at slower rates than another recombinant human monoclonal IgG4. While F(ab')2 fragments were rapidly cleared from the circulation, thus leading to a rapid loss of emicizumab procoagulant activity, low amounts of single-cleaved intermediate IgG persisted for several days. Moreover, the IdeS-mediated elimination of the neutralizing anti-FVIII IgG and restoration of the hemostatic efficacy of exogenous FVIII were not impaired by the presence of emicizumab and polyclonal human IgG in inhibitor-positive HA mice. CONCLUSION: Our results suggest that IdeS could be administered to inhibitor-positive patients with HA receiving emicizumab prophylaxis to improve and ease the management of breakthrough bleeds or programmed major surgeries.


Asunto(s)
Anticuerpos Biespecíficos , Hemofilia A , Humanos , Animales , Ratones , Hemofilia A/tratamiento farmacológico , Factor VIII/uso terapéutico , Anticuerpos Biespecíficos/uso terapéutico , Hemorragia/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Inmunoglobulina G
3.
J Thromb Haemost ; 21(9): 2405-2417, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37271431

RESUMEN

BACKGROUND: Transplacental delivery of maternal immunoglobulin G (IgG) provides humoral protection during the first months of life until the newborn's immune system reaches maturity. The maternofetal interface has been exploited therapeutically to replace missing enzymes in the fetus, as shown in experimental mucopolysaccharidoses, or to shape adaptive immune repertoires during fetal development and induce tolerance to self-antigens or immunogenic therapeutic molecules. OBJECTIVES: To investigate whether proteins that are administered to pregnant mice or endogenously present in their circulation may be delivered through the placenta. METHODS: We engineered monovalent immunoglobulin G (FabFc) specific for different domains of human factor VIII (FVIII), a therapeutically relevant model antigen. FabFc was injected with exogenous FVIII into pregnant severe hemophilia A mice or pregnant mice expressing human FVIII following AAV8-mediated gene therapy. FabFc and FVIII were detected in the pregnant mice and/or fetuses by enzyme-linked immunosorbent assay and immunohistochemistry. RESULTS: Administration of FabFc to pregnant mice allowed the maternofetal delivery of FVIII in a FcRn-dependent manner. FVIII antigen levels achieved in the fetuses represented 10% of normal plasma levels in the human. We identified antigen/FabFc complex stability, antigen size, and shielding of promiscuous protein patches as key parameters to foster optimal antigen delivery. CONCLUSION: Our results pave the way toward the development of novel strategies for the in utero delivery of endogenous maternal proteins to replace genetically deficient fetal proteins or to educate the immune system and favor active immune tolerance upon protein encounter later in life.


Asunto(s)
Hemofilia A , Inmunoglobulina G , Embarazo , Femenino , Ratones , Humanos , Animales , Factor VIII , Hemofilia A/genética , Hemofilia A/terapia , Placenta , Terapia Genética , Tolerancia Inmunológica
4.
Haematologica ; 108(5): 1322-1334, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36655430

RESUMEN

Neutralizing anti-factor VIII (FVIII) antibodies, known as FVIII inhibitors, represent a major drawback of replacement therapy in persons with congenital hemophilia A (PwHA), rendering further infusions of FVIII ineffective. FVIII inhibitors can also appear in non-hemophilic individuals causing acquired hemophilia A (AHA). The use of non-FVIII bypassing agents in cases of bleeds or surgery in inhibitor-positive patients is complicated by the lack of reliable biological monitoring and increased thrombotic risk. Imlifidase (IdeS) is an endopeptidase that degrades human immunoglobulin G (IgG); it was recently approved for hyperimmune patients undergoing renal transplants. Here we investigated the ability of IdeS to eliminate FVIII inhibitors in vitro and in a model of inhibitor-positive HA mice. IdeS cleaved anti-FVIII plasma IgG from PwHA and AHA patients, and hydrolyzed recombinant human anti-FVIII IgG independently from their subclass or specificity for the A2, A3, C1 or C2 domains of FVIII. In HA mice passively immunized with recombinant human anti-FVIII IgG, IdeS restored the hemostatic efficacy of FVIII, as evidenced by the correction of the bleeding tendency. Our results provide the proof of concept for the transient removal of FVIII inhibitors by IdeS, thereby opening a therapeutic window for efficient FVIII replacement therapy in inhibitor-positive patients.


Asunto(s)
Hemofilia A , Hemostáticos , Humanos , Ratones , Animales , Hemofilia A/tratamiento farmacológico , Hemorragia , Inmunoglobulina G , Inmunosupresores/uso terapéutico
6.
Cell Immunol ; 355: 104151, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32615414

RESUMEN

B cells with regulatory properties (Bregs) were identified in human and in mice among different B-cell subsets. Their regulatory properties rely mainly on the production of anti-inflammatory cytokines, in particular IL10, IL-35 and TGFß, and were extensively studied in mouse models of autoimmune and inflammatory diseases. However, the exact nature of the stimulatory signals conferring regulatory properties to B cells is still not clear. We serendipitously observed that fluorescein isothiocyanate (FITC) binds to a significant proportion of naïve mouse B cells. Binding of FITC to the B-cell surface implicated at least in part the B-cell receptor. It triggered IL-10 production and allowed the endocytosis of FITC-coupled antigens followed by their presentation to CD4+ T cells. In particular, B cells incubated with FITC-OVA polarized OTII T cells towards a Tr1/Th2 phenotype in vitro. Further, the adoptive transfer of B cells incubated with FITC-labeled myelin oligodendrocyte glycoprotein peptide protected mice from experimental autoimmune encephalomyelitis, a T-cell-dependent autoimmune model. Together, the data show that FITC-stimulated B cells polarize immune responses towards Tr1/Th2 and acquire immuno-modulatory properties.


Asunto(s)
Linfocitos B Reguladores/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Animales , Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Linfocitos B Reguladores/inmunología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Fluoresceína/metabolismo , Fluoresceína/farmacología , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceína-5-Isotiocianato/farmacología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Interleucinas/inmunología , Interleucinas/metabolismo , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo
7.
Front Immunol ; 11: 810, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477339

RESUMEN

In humans, maternal IgGs are transferred to the fetus from the second trimester of pregnancy onwards. The transplacental delivery of maternal IgG is mediated by its binding to the neonatal Fc receptor (FcRn) after endocytosis by the syncytiotrophoblast. IgGs present in the maternal milk are also transferred to the newborn through the digestive epithelium upon binding to the FcRn. Importantly, the binding of IgGs to the FcRn is also responsible for the recycling of circulating IgGs that confers them with a long half-life. Maternally delivered IgG provides passive immunity to the newborn, for instance by conferring protective anti-flu or anti-pertussis toxin IgGs. It may, however, lead to the development of autoimmune manifestations when pathological autoantibodies from the mother cross the placenta and reach the circulation of the fetus. In recent years, strategies that exploit the transplacental delivery of antigen/IgG complexes or of Fc-fused proteins have been validated in mouse models of human diseases to impose antigen-specific tolerance, particularly in the case of Fc-fused factor VIII (FVIII) domains in hemophilia A mice or pre-pro-insulin (PPI) in the case of preclinical models of type 1 diabetes (T1D). The present review summarizes the mechanisms underlying the FcRn-mediated transcytosis of IgGs, the physiopathological relevance of this phenomenon, and the repercussion for drug delivery and shaping of the immune system during its ontogeny.


Asunto(s)
Antígenos/inmunología , Tolerancia Inmunológica , Intercambio Materno-Fetal/inmunología , Animales , Autoanticuerpos/metabolismo , Femenino , Feto/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Sistema Inmunológico/embriología , Sistema Inmunológico/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Placenta/inmunología , Embarazo , Transporte de Proteínas/inmunología , Receptores Fc/metabolismo , Transcitosis/inmunología
8.
Nat Med ; 26(7): 1096-1101, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32483358

RESUMEN

Neutralizing antibodies to adeno-associated virus (AAV) vectors are highly prevalent in humans1,2, and block liver transduction3-5 and vector readministration6; thus, they represent a major limitation to in vivo gene therapy. Strategies aimed at overcoming anti-AAV antibodies are being studied7, which often involve immunosuppression and are not efficient in removing pre-existing antibodies. Imlifidase (IdeS) is an endopeptidase able to degrade circulating IgG that is currently being tested in transplant patients8. Here, we studied if IdeS could eliminate anti-AAV antibodies in the context of gene therapy. We showed efficient cleavage of pooled human IgG (intravenous Ig) in vitro upon endopeptidase treatment. In mice passively immunized with intravenous Ig, IdeS administration decreased anti-AAV antibodies and enabled efficient liver gene transfer. The approach was scaled up to nonhuman primates, a natural host for wild-type AAV. IdeS treatment before AAV vector infusion was safe and resulted in enhanced liver transduction, even in the setting of vector readministration. Finally, IdeS reduced anti-AAV antibody levels from human plasma samples in vitro, including plasma from prospective gene therapy trial participants. These results provide a potential solution to overcome pre-existing antibodies to AAV-based gene therapy.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Dependovirus/genética , Terapia Genética , Vectores Genéticos/efectos adversos , Animales , Anticuerpos Antiidiotipos/genética , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/inmunología , Cápside/inmunología , Dependovirus/inmunología , Endopeptidasas/inmunología , Vectores Genéticos/uso terapéutico , Humanos , Inmunoglobulina G/farmacología , Hígado/inmunología , Hígado/metabolismo , Ratones
9.
Med Sci (Paris) ; 36(4): 341-347, 2020 Apr.
Artículo en Francés | MEDLINE | ID: mdl-32356710

RESUMEN

The use of therapeutic proteins induces in some patients the appearance of neutralizing antibodies. This is the case of pro-coagulant factor VIII (FVIII) used in patients with hemophilia A. Several parameters related to the protein itself, to the type of pathology or to the patients, condition the immunogenicity of a therapeutic protein. Understanding these parameters would help to anticipate or prevent the development of neutralizing antibodies. In the case of FVIII, we propose that the development of neutralizing antibodies does not result from an unpredicted immune response but rather from the inability of the patient's organism to develop an anti-inflammatory or regulatory response.


TITLE: Origine et nature de la réponse immunitaire neutralisante contre le facteur VIII thérapeutique. ABSTRACT: L'utilisation de protéines thérapeutiques se heurte, chez certains patients, à l'apparition d'anticorps neutralisants. C'est le cas, par exemple, du facteur VIII pro-coagulant qui est utilisé pour traiter les patients atteints d'hémophilie A. Plusieurs paramètres, liés à la protéine elle-même, au type de pathologie ou aux patients, conditionnent l'immunogénicité d'une protéine thérapeutique. Les comprendre permettrait d'anticiper ou de prévenir la survenue d'anticorps neutralisants. Nous proposons dans cette revue de montrer que, dans le cas du facteur VIII, la survenue de ces anticorps neutralisants ne résulte pas d'une réponse immunitaire inopinée, mais plutôt de l'incapacité de l'organisme des patients à développer une réponse anti-inflammatoire ou régulatrice.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Factor VIII/inmunología , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemofilia A/terapia , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos/fisiología , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica/fisiología
10.
Front Immunol ; 11: 393, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32273875

RESUMEN

The development of an immune response against therapeutic factor VIII is the major complication in hemophilia A patients. Oligomannose carbohydrates at N239 and/or N2118 on factor VIII allow its binding to the macrophage mannose receptor expressed on human dendritic cells, thereby leading to factor VIII endocytosis and presentation to CD4+ T lymphocytes. Here, we investigated whether altering the interaction of factor VIII with mannose-sensitive receptors on antigen-presenting cells may be a strategy to reduce factor VIII immunogenicity. Gene transfer experiments in factor VIII-deficient mice indicated that N239Q and/or N2118Q factor VIII mutants have similar specific activities as compared to non-mutated factor VIII; N239Q/N2118Q mutant corrected blood loss upon tail clip. Production of the corresponding recombinant FVIII mutants or light chains indicated that removal of the N-linked glycosylation site at N2118 is sufficient to abrogate in vitro the activation of FVIII-specific CD4+ T cells by human monocyte-derived dendritic cells. However, removal of mannose-ending glycans at N2118 did not alter factor VIII endocytosis and presentation to CD4+ T cells by mouse antigen-presenting cells. In agreement with this, the N2118Q mutation did not reduce factor VIII immunogenicity in factor VIII-deficient mice. Our results highlight differences in the endocytic pathways between human and mouse dendritic cell subsets, and dissimilarities in tissue distribution and function of endocytic receptors such as CD206 in both species. Further investigations in preclinical models of hemophilia A closer to humans are needed to decipher the exact role of mannose-ending glycans in factor VIII immunogenicity.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Factor VIII/inmunología , Activación de Linfocitos/inmunología , Animales , Factor VIII/química , Factor VIII/genética , Humanos , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Manosa/química , Manosa/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/inmunología , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/inmunología , Mutación , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo
11.
Commun Biol ; 3(1): 96, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32132640

RESUMEN

Therapeutic normal IgG intravenous immunoglobulin (IVIG) is a well-established first-line immunotherapy for many autoimmune and inflammatory diseases. Though several mechanisms have been proposed for the anti-inflammatory actions of IVIG, associated signaling pathways are not well studied. As ß-catenin, the central component of the canonical Wnt pathway, plays an important role in imparting tolerogenic properties to dendritic cells (DCs) and in reducing inflammation, we explored whether IVIG induces the ß-catenin pathway to exert anti-inflammatory effects. We show that IVIG in an IgG-sialylation independent manner activates ß-catenin in human DCs along with upregulation of Wnt5a secretion. Mechanistically, ß-catenin activation by IVIG requires intact IgG and LRP5/6 co-receptors, but FcγRIIA and Syk are not implicated. Despite induction of ß-catenin, this pathway is dispensable for anti-inflammatory actions of IVIG in vitro and for mediating the protection against experimental autoimmune encephalomyelitis in vivo in mice, and reciprocal regulation of effector Th17/Th1 and regulatory T cells.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , beta Catenina/metabolismo , Animales , Antiinflamatorios/farmacología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/efectos de los fármacos
12.
Front Immunol ; 11: 558811, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424828

RESUMEN

We report the observation of a 75-year-old patient referred for cervical lymphadenopathies. A pre-lymphadenectomy blood work revealed an asymptomatic elevation of aPTT with low factor VIII (FVIII) levels and high anti-FVIII antibodies titers, consistent with acquired hemophilia A (AHA). Histological work-up of a cervical lymphadenopathy revealed benign follicular hyperplasia with IgG4+ lymphoplasmacytic infiltration; and serum IgG4 levels were markedly elevated, compatible with IgG4-related disease (IgG4-RD). He was successfully treated with a 9-month course of prednisone, secondarily associated with rituximab when an AHA relapse occurred. As this patient presented with an unusual association of rare diseases, we wondered whether there was a link between the two conditions. Our first hypothesis was that the anti-FVIII autoantibodies could be directly produced by the proliferating IgG4+ plasma cells as a result of broken tolerance to autologous FVIII. To test this assumption, we determined the anti-FVIII IgG subclasses in our patient and in a control group of 11 AHA patients without IgG4-RD. The FVIII inhibitor was mostly IgG4, with an anti-FVIII IgG4/IgG1 ratio of 42 at diagnosis and 268 at relapse in our patient; similar values were observed in non-IgG4-RD AHA patients. As a second hypothesis, we considered whether the anti-FVIII activity could be the result of a non-specific autoantibody production due to polyclonal IgG4+ plasma cell proliferation. To test this hypothesis, we measured the anti-FVIII IgG4/total IgG4 ratio in our patient, as well as in several control groups: 11 AHA patients without IgG4-RD, 8 IgG4-RD patients without AHA, and 11 healthy controls. We found that the median [min-max] ratio was higher in AHA-only controls (2.4 10-2 [5.7 10-4-1.79 10-1]), an oligoclonal setting in which only anti-FVIII plasma cells proliferate, than in IgG4-RD-only controls (3.0 10-5 [2.0 10-5-6.0 10-5]), a polyclonal setting in which all IgG4+ plasma cells proliferate equally. Our patient had intermediate ratio values (2.7 10-3 at diagnosis and 1.0 10-3 at relapse), which could plead for a combination of both mechanisms. Although no definitive conclusion can be drawn, we hypothesized that the anti-FVIII autoantibody production in our IgG4-RD AHA patient could be the result of both broken tolerance to FVIII and bystander polyclonal IgG4+ plasma cell proliferation.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Hemofilia A/diagnóstico , Hemofilia A/etiología , Enfermedad Relacionada con Inmunoglobulina G4/complicaciones , Enfermedad Relacionada con Inmunoglobulina G4/inmunología , Anciano , Autoanticuerpos/inmunología , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Factor VIII/inmunología , Femenino , Hemofilia A/sangre , Hemofilia A/terapia , Humanos , Inmunoglobulina G/inmunología , Inmunohistoquímica , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Masculino , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
13.
Haematologica ; 105(4): 1129-1137, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31289204

RESUMEN

The treatment or prevention of bleeding in patients with hemophilia A relies on replacement therapy with different factor VIII (FVIII)-containing products or on the use of by-passing agents, i.e., activated prothrombin complex concentrates or recombinant activated factor VII. Emerging approaches include the use of bispecific anti-factor IXa/factor X antibodies, anti-tissue factor pathway inhibitor antibodies, interfering RNA to antithrombin, and activated protein C-specific serpins or gene therapy. The latter strategies are, however, hampered by the short clinical experience and potential adverse effects including the absence of tight temporal and spatial control of coagulation and the risk of uncontrolled insertional mutagenesis. Systemic delivery of mRNA allows endogenous production of the corresponding encoded protein. Thus, injection of erythropoietin-encoding mRNA in a lipid nanoparticle formulation resulted in increased erythropoiesis in mice and macaques. Here, we demonstrate that a single injection of in vitro transcribed B domain-deleted FVIII-encoding mRNA to FVIII-deficient mice enables endogenous production of pro-coagulant FVIII. Circulating FVIII:C levels above 5% of normal levels were maintained for up to 72 h, with an estimated half-life of FVIII production of 17.9 h, and corrected the bleeding phenotype in a tail clipping assay. The endogenously produced FVIII did however exhibit low specific activity and induced a potent neutralizing IgG response upon repeated administration of the mRNA. Our results suggest that the administration of mRNA is a plausible strategy for the endogenous production of proteins characterized by poor translational efficacy. The use of alternative mRNA delivery systems and improved FVIII-encoding mRNA should foster the production of functional molecules and reduce their immunogenicity.


Asunto(s)
Anticuerpos Biespecíficos , Hemofilia A , Animales , Factor VIII/genética , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemorragia/terapia , Humanos , Ratones , ARN Mensajero/genética
14.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1152-1159, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30684639

RESUMEN

The nitrone spin trap 5,5­dimethyl­1­pyrroline N­oxide (DMPO) dampens endotoxin-induced and TLR4-driven priming of macrophages, but the mechanism remains unknown. The available information suggests a direct binding of DMPO to the TIR domain, which is shared between TLRs. However, TLR2-TIR domain is the only TLR that have been crystallized. Our in silico data show that DMPO binds to four specific residues in the BB-loop within the TLR2-TIR domain. Our functional analysis using hTLR2.6-expressing HEKs cells showed that DMPO can block zymosan-triggered-TLR2-mediated NF-κB activation. However, DMPO did not affect the overall TLR2-MyD88 protein-protein interaction. DMPO binds to the BB-loop in the TIR-domain and dampens downstream signaling without affecting the overall TIR-MyD88 interaction. These data encourage the use of DMPO-derivatives as potential mechanism-based inhibitors of TLR-triggered inflammation.


Asunto(s)
Óxidos N-Cíclicos/metabolismo , Inflamación/metabolismo , Óxidos de Nitrógeno/metabolismo , Transducción de Señal , Marcadores de Spin , Receptor Toll-Like 2/metabolismo , Animales , Óxidos N-Cíclicos/química , Células HEK293 , Humanos , Inflamación/inmunología , Ratones , Simulación de Dinámica Molecular , Factor 88 de Diferenciación Mieloide/química , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/química , FN-kappa B/metabolismo , Óxidos de Nitrógeno/química , Unión Proteica , Dominios Proteicos , Células RAW 264.7 , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/química
15.
Haematologica ; 104(5): 1046-1054, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30545924

RESUMEN

Hemophilia A is a rare hemorrhagic disorder caused by the lack of functional pro-coagulant factor VIII. Factor VIII replacement therapy in patients with severe hemophilia A results in the development of inhibitory anti-factor VIII IgG in up to 30% of cases. To date, immune tolerance induction, with daily injection of large amounts of factor VIII, is the only strategy to eradicate factor VIII inhibitors. This strategy is, however, efficient in only 60-80% of patients. We investigated whether blocking B-cell receptor signaling upon inhibition of Bruton tyrosine kinase prevents anti-factor VIII immune responses in a mouse model of severe hemophilia A. Factor VIII-naïve and factor VIII-sensitized factor VIII-deficient mice were fed with the selective inhibitor of Bruton tyrosine kinase, (R)-5-amino-1-(1-cyanopiperidin-3-yl)-3-(4-[2,4-difluorophenoxyl] phenyl)-1H pyrazole-4-carboxamide (PF-06250112), to inhibit B-cell receptor signaling prior to challenge with exogenous factor VIII. The consequences on the anti-factor VIII immune response were studied. Inhibition of Bruton tyrosine kinase during the primary anti-factor VIII immune response in factor VIII-naïve mice did not prevent the development of inhibitory anti-factor VIII IgG. In contrast, the anti-factor VIII memory B-cell response was consistently reduced upon treatment of factor VIII-sensitized mice with the Bruton tyrosine kinase inhibitor. The Bruton tyrosine kinase inhibitor reduced the differentiation of memory B cells ex vivo and in vivo following adoptive transfer to factor VIII-naïve animals. Taken together, our data identify inhibition of Bruton tyrosine kinase using PF-06250112 as a strategy to limit the reactivation of factor VIII-specific memory B cells upon re-challenge with therapeutic factor VIII.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Factor VIII/fisiología , Hemofilia A/inmunología , Memoria Inmunológica/inmunología , Piperidinas/farmacología , Pirazoles/farmacología , Animales , Formación de Anticuerpos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Factor VIII/administración & dosificación , Factor VIII/antagonistas & inhibidores , Hemofilia A/tratamiento farmacológico , Hemofilia A/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Inmunoglobulina G/efectos de los fármacos , Inmunoglobulina G/inmunología , Memoria Inmunológica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Semin Thromb Hemost ; 44(6): 517-530, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29864775

RESUMEN

The immunogenicity of therapeutic factor VIII (FVIII) in patients with hemophilia A has been puzzling scientific and clinical communities for more than 3 decades. Indeed, the development of inhibitory antibodies to FVIII remains a major clinical challenge and is associated with enormous societal costs. Thus, the reasons for which a presumably innocuous, short-lived, intravenously administered glycoprotein triggers such a deleterious, long-lasting neutralizing immune response is an enigma. This review does not pretend to bring an answer to this challenging question. It will however summarize the latest findings regarding the molecular interactions at play in the recognition of FVIII by the immune cells, the validity of the proposed risk factors for FVIII alloimmunization, and the different solutions that allow induction of FVIII-specific tolerance in preclinical models of hemophilia A.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/inmunología , Humanos
17.
Cell Immunol ; 331: 22-29, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29751951

RESUMEN

Hemophilia A is a X-linked recessive bleeding disorder consecutive to the lack of circulating pro-coagulant factor VIII (FVIII). The most efficient strategy to treat or prevent bleeding in patients with hemophilia A relies on replacement therapy using exogenous FVIII. Commercially available recombinant FVIII are produced using an expensive perfusion technology in stainless steel fermenters. A fed-batch fermentation technology was recently developed to produce 'Neureight', a full-length recombinant human FVIII, in Chinese hamster ovary (CHO) cells. Here, we investigated the structural and functional integrity and lack of increased immunogenicity of Neureight, as compared to two commercially available full-length FVIII products, Helixate and Advate, produced in baby hamster kidney or CHO cells, respectively. Our results demonstrate the purity, stability and functional integrity of Neureight with a standard specific activity of 4235 ±â€¯556 IU/mg. The glycosylation and sulfation profiles of Neureight were similar to that of Advate, with the absence of the antigenic carbohydrate epitopes α-Gal and Neu5Gc, and with sulfation of Y1680, that is critical for FVIII binding to von Willebrand factor (VWF). The endocytosis of Neureight by human immature dendritic cells was inhibited by VWF, and its half-life in FVIII-deficient mice was similar to that of Advate, confirming unaltered binding to VWF. In vitro and in vivo assays indicated a similar immunogenicity for Neureight, Advate and Helixate. In conclusion, the production of full-length FVIII in a fed-batch fermentation mode generates a product that presents similar biochemical, functional and immunogenic properties as products developed using the classical perfusion technology.


Asunto(s)
Reactores Biológicos , Factor VIII/inmunología , Hemofilia A/inmunología , Proteínas Recombinantes/inmunología , Animales , Células CHO , Cricetinae , Cricetulus , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Endocitosis/inmunología , Factor VIII/genética , Factor VIII/metabolismo , Fermentación , Hemofilia A/tratamiento farmacológico , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico
18.
JCI Insight ; 3(6)2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29563336

RESUMEN

Hemostatic defects are treated using coagulation factors; however, clot formation also requires a procoagulant phospholipid (PL) surface. Here, we show that innate immune cell-derived enzymatically oxidized phospholipids (eoxPL) termed hydroxyeicosatetraenoic acid-phospholipids (HETE-PLs) restore hemostasis in human and murine conditions of pathological bleeding. HETE-PLs abolished blood loss in murine hemophilia A and enhanced coagulation in factor VIII- (FVIII-), FIX-, and FX-deficient human plasma . HETE-PLs were decreased in platelets from patients after cardiopulmonary bypass (CPB). To explore molecular mechanisms, the ability of eoxPL to stimulate individual isolated coagulation factor/cofactor complexes was tested in vitro. Extrinsic tenase (FVIIa/tissue factor [TF]), intrinsic tenase (FVIIIa/FIXa), and prothrombinase (FVa/FXa) all were enhanced by both HETE-PEs and HETE-PCs, suggesting a common mechanism involving the fatty acid moiety. In plasma, 9-, 15-, and 12-HETE-PLs were more effective than 5-, 11-, or 8-HETE-PLs, indicating positional isomer specificity. Coagulation was enhanced at lower lipid/factor ratios, consistent with a more concentrated area for protein binding. Surface plasmon resonance confirmed binding of FII and FX to HETE-PEs. HETE-PEs increased membrane curvature and thickness, but not surface charge or homogeneity, possibly suggesting increased accessibility to cations/factors. In summary, innate immune-derived eoxPL enhance calcium-dependent coagulation factor function, and their potential utility in bleeding disorders is proposed.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Hemorragia/enzimología , Hemorragia/metabolismo , Fosfolípidos/metabolismo , Trombina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Coagulación Sanguínea , Factores de Coagulación Sanguínea/genética , Plaquetas , Puente Cardiopulmonar/efectos adversos , Proteínas Portadoras , Cisteína Endopeptidasas , Factor IX/genética , Factor VIII/genética , Factor VIIa/metabolismo , Factor X/genética , Hemofilia A , Hemorragia/prevención & control , Hemostasis , Humanos , Ácidos Hidroxieicosatetraenoicos , Lipoproteínas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas de Neoplasias , Resonancia por Plasmón de Superficie , Tromboplastina/antagonistas & inhibidores , Tromboplastina/metabolismo
19.
Mol Immunol ; 97: 1-7, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29525557

RESUMEN

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. Humanized CVF (hCVF) is a human C3 derivative where the C-terminal 168 amino acid residues were replaced with the homologous sequence from CVF. hCVF has been shown in multiple models of disease with complement pathology to be a promising therapeutic agent, with no observed adverse effects. Here we describe the antibody response to hCVF in two different strains of mice. hCVF was able to repeatedly decomplement the mice after four injections in weekly intervals, demonstrating the absence of a neutralizing antibody response. In contrast, natural CVF caused decomplementation in all mice only after the first administration. After two additional administrations of natural CVF, decomplementation was inconsistent and varied tremendously from mouse to mouse. After the fourth administration, natural CVF was essentially unable to deplete complement, consistent with the known generation of a neutralizing antibody response. We also analyzed the IgG antibody response to hCVF. There was great variation, with approximately one quarter of the mice exhibiting non-detectable levels of anti-hCVF IgG, and another quarter very low levels. The levels of anti-hCVF IgG did not correlate with the levels of remaining C3. The anti-hCVF antibodies cross-reacted with natural CVF, recombinant CVF, and human C3. Whereas overall the level of anti-hCVF IgG cross-reacting with human C3 was lower compared to rCVF or nCVF, mice with higher levels of anti-hCVF IgG exhibited higher binding to CVF and human C3, excluding the possibility that higher antibody levels reflect preferential immunogenicity of CVF-specific or human C3-specific epitopes.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Formación de Anticuerpos , Venenos Elapídicos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Drosophila melanogaster , Venenos Elapídicos/química , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/química
20.
Cell Immunol ; 325: 64-68, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29395036

RESUMEN

The development of antibodies against therapeutic factor VIII (FVIII) represents the major complication of replacement therapy in patients with severe hemophilia A. Amongst the environmental risk factors that influence the anti-FVIII immune response, the presence of active bleeding or hemarthrosis has been evoked. Endothelium damage is typically associated with the release of oxidative compounds. Here, we addressed whether oxidation contributes to FVIII immunogenicity. The control with N-acetyl cysteine of the oxidative status in FVIII-deficient mice, a model of severe hemophilia A, reduced the immune response to exogenous FVIII. Ex vivo exposure of therapeutic FVIII to HOCl induced a mild oxidation of the molecule as evidenced by the loss of free amines and resulted in increased FVIII immunogenicity in vivo when compared to native FVIII. The increased immunogenicity of oxidized FVIII was not reverted by treatment of mice with N-acetyl cysteine, and did not implicate an increased maturation of professional antigen-presenting cells. Our data document that oxidation influences the immunogenicity of therapeutic FVIII.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/inmunología , Hemofilia A/metabolismo , Acetilcisteína/farmacología , Animales , Anticuerpos/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Modelos Animales de Enfermedad , Factor VIII/metabolismo , Factor VIII/farmacología , Hemofilia A/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA