Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Blood ; 129(24): 3147-3154, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28432221

RESUMO

In hemophilia A, the most severe complication of factor VIII (FVIII) replacement therapy involves the formation of FVIII neutralizing antibodies, also known as inhibitors, in 25% to 30% of patients. This adverse event is associated with a significant increase in morbidity and economic burden, thus highlighting the need to identify methods to limit FVIII immunogenicity. Inhibitor development is regulated by a complex balance of genetic factors, such as FVIII genotype, and environmental variables, such as coexistent inflammation. One of the hypothesized risk factors of inhibitor development is the source of the FVIII concentrate, which could be either recombinant or plasma derived. Differential immunogenicity of these concentrates has been documented in several recent epidemiologic studies, thus generating significant debate within the hemophilia treatment community. To date, these discussions have been unable to reach a consensus regarding how these outcomes might be integrated into enhancing clinical care. Moreover, the biological mechanistic explanations for the observed differences are poorly understood. In this article, we complement the existing epidemiologic investigations with an overview of the range of possible biochemical and immunologic mechanisms that may contribute to the different immune outcomes observed with plasma-derived and recombinant FVIII products.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fator VIII , Hemofilia A , Plasma , Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Humanos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
2.
Blood ; 127(26): 3439-49, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27034428

RESUMO

Inflammatory signals such as pathogen- and danger-associated molecular patterns have been hypothesized as risk factors for the initiation of the anti-factor VIII (FVIII) immune response seen in 25% to 30% of patients with severe hemophilia A (HA). In these young patients, vaccines may be coincidentally administered in close proximity with initial exposure to FVIII, thereby providing a source of such stimuli. Here, we investigated the effects of 3 vaccines commonly used in pediatric patients on FVIII immunogenicity in a humanized HA murine model with variable tolerance to recombinant human FVIII (rhFVIII). Mice vaccinated intramuscularly against the influenza vaccine prior to multiple infusions of rhFVIII exhibited a decreased incidence of rhFVIII-specific neutralizing and nonneutralizing antibodies. Similar findings were observed with the addition of an adjuvant. Upon exposure to media from influenza- or FVIII-stimulated lymph node or splenic lymphocytes, naïve CD4(+) lymphocytes preferentially migrated toward media from influenza-stimulated cells, indicating that antigen competition, by means of lymphocyte recruitment to the immunization site, is a potential mechanism for the observed decrease in FVIII immunogenicity. We also observed no differences in incidence or titer of rhFVIII-specific antibodies and inhibitors in mice exposed to the live-attenuated measles-mumps-rubella vaccine regardless of route of administration. Together, our results suggest that concomitant FVIII exposure and vaccination against influenza does not increase the risk of inhibitor formation and may in fact decrease anti-FVIII immune responses.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Autoanticorpos/imunologia , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fator VIII/antagonistas & inibidores , Hemofilia A/imunologia , Vacinas contra Influenza/farmacologia , Vacinação , Animais , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Fator VIII/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout
3.
Haematologica ; 103(11): 1925-1936, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30002126

RESUMO

Immune responses to factor VIII remain the greatest complication in the treatment of severe hemophilia A. Recent epidemiological evidence has highlighted that recombinant factor VIII produced in baby hamster kidney cells is more immunogenic than factor VIII produced in Chinese hamster ovary cells. Glycosylation differences have been hypothesized to influence the immunogenicity of these synthetic concentrates. In two hemophilia A mouse models, baby hamster kidney cell-derived factor VIII elicited a stronger immune response compared to Chinese hamster ovary cell-derived factor VIII. Furthermore, factor VIII produced in baby hamster kidney cells exhibited accelerated clearance from circulation independent of von Willebrand factor. Lectin and mass spectrometry analysis of total N-linked glycans revealed differences in high-mannose glycans, sialylation, and the occupancy of glycan sites. Factor VIII desialylation did not influence binding to murine splenocytes or dendritic cells, nor surface co-stimulatory molecule expression. We did, however, observe increased levels of immunoglobulin M specific to baby hamster kidney-derived factor VIII in naïve hemophilia A mice. De-N-glycosylation enhanced immunoglobulin M binding, suggesting that N-glycan occupancy masks epitopes. Elevated levels of immunoglobulin M and immunoglobulin G specific to baby hamster kidney-derived factor VIII were also observed in healthy individuals, and de-N-glycosylation increased immunoglobulin G binding. Collectively, our data suggest that factor VIII produced in baby hamster kidney cells is more immunogenic than that produced in Chinese hamster ovary cells, and that incomplete occupancy of N-linked glycosylation sites leads to the formation of immunoglobulin M- and immunoglobulin G-factor VIII immune complexes that contribute to the enhanced clearance and immunogenicity in these mouse models of hemophilia A.


Assuntos
Fator VIII , Hemofilia A , Animais , Células CHO , Cricetulus , Modelos Animais de Doenças , Fator VIII/imunologia , Fator VIII/farmacologia , Feminino , Glicosilação , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/patologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia
4.
Haematologica ; 103(8): 1403-1413, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29674503

RESUMO

The development of inhibitory antibodies to factor VIII is the most serious complication of replacement therapy in hemophilia A. Activation of the innate immune system during exposure to this protein contributes to inhibitor development. However, avoidance of factor VIII exposure during innate immune system activation by external stimuli (e.g., vaccines) has not been consistently shown to prevent inhibitors. We hypothesized that dexamethasone, a drug with potent anti-inflammatory effects, could prevent inhibitors by promoting immunologic tolerance to factor VIII in hemophilia A mice. Transient dexamethasone treatment during ainitial factor VIII exposure reduced the incidence of anti-factor VIII immunoglobulin G in both a conventional hemophilia A mouse model (E16KO, 77% vs 100%, P=0.048) and a hemophilia A mouse model with a humanized major histocompatibility complex type II transgene (E17KO/hMHC, 6% vs 33%, P=0.0048). More importantly, among E17KO/hMHC mice that did not develop anti-factor VIII immunoglobulin G after initial exposure, dexamethasone-treated mice were less likely to develop a response after re-exposure six (7% vs 52%, P=0.005) and 16 weeks later (7% vs 50%, P=0.097). Similar results were obtained even when factor VIII re-exposure occurred in the context of lipopolysaccharide (30% vs 100%, P=0.069). The ability of these mice to develop immunoglobulin G to human von Willebrand factor, a structurally unrelated antigen, remained unaffected by treatment. Transient dexamethasone administration therefore promotes antigen-specific immunologic tolerance to factor VIII. This effect is associated with an increase in the percentage of thymic regulatory T cells (12.06% vs 4.73%, P<0.001) and changes in the thymic messenger ribonucleic acid transcription profile.


Assuntos
Dexametasona/uso terapêutico , Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Tolerância Imunológica , Animais , Anticorpos , Modelos Animais de Doenças , Imunidade Inata , Imunoglobulina G , Camundongos , RNA Mensageiro/análise , Linfócitos T Reguladores/imunologia , Timo/imunologia , Fatores de Tempo , Transcrição Gênica
5.
Cell Immunol ; 301: 82-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26547364

RESUMO

The enigma that is factor VIII immunogenicity remains ever pertinent in the treatment of hemophilia A. Development of neutralizing antibodies against the therapeutic protein in 25-30% of patients likely depends on the appropriate activation of the innate immune response shortly following antigen encounter. Our understanding of this important immunological synapse remains ill-defined. In this review, we examine the three distinct factors contributing to the fate of factor VIII almost immediately after infusion: the characteristics of the protein, the cell, and the microenvironment. We propose a continuum between clearance and antigen presentation that facilitates removal of FVIII from circulation leading to either tolerance or immunity.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica/imunologia , Imunidade Inata/imunologia , Anticorpos Neutralizantes/imunologia , Apresentação de Antígeno/imunologia , Humanos
6.
Cell Immunol ; 301: 2-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26676073

RESUMO

The development of neutralizing anti-factor VIII (FVIII) antibodies (inhibitors) remains a major challenge for FVIII replacement therapy in hemophilia A patients. The adaptive immune response plays a crucial role in the development and maintenance of inhibitors. In this review, we focus on our current understanding of FVIII interactions with cells of the adaptive immune system and the phenotype of the resultant response. Additionally, we examine both current and novel FVIII tolerance induction methods that function at the level of the adaptive immune response.


Assuntos
Imunidade Adaptativa/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Anticorpos Neutralizantes/imunologia , Humanos
7.
Blood ; 123(26): 4045-53, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24829206

RESUMO

Ex vivo gene therapy strategies avoid systemic delivery of viruses thereby mitigating the risk of vector-associated immunogenicity. Previously, we delivered autologous factor VIII (FVIII)-expressing blood outgrowth endothelial cells (BOECs) to hemophilia A mice and showed that these cells remained sequestered within the implanted matrix and provided therapeutic levels of FVIII. Prior to translating this strategy into the canine (c) model of hemophilia A, we increased cFVIII transgene expression by at least 100-fold with the use of the elongation factor 1 alpha (EF1α) promoter and a strong endothelial enhancer element. BOECs isolated from hemophilia A dogs transduced with this lentiviral vector express levels of cFVIII ranging between 1.0 and 1.5 U/mL per 10(6) cells over 24 hours. Autologous BOECs have been implanted into the omentum of 2 normal and 3 hemophilia A dogs. These implanted cells formed new vessels in the omentum. All 3 hemophilia A dogs treated with FVIII-expressing autologous BOECs developed anti-FVIII immunoglobulin G2 antibodies, but in only 2 of the dogs were these antibodies inhibitory. FVIII antigen levels >40% in the absence of FVIII coagulant function were detected in the circulation for up to a year after a single gene therapy treatment, indicating prolonged cellular viability and synthesis of FVIII.


Assuntos
Autoanticorpos , Inibidores dos Fatores de Coagulação Sanguínea , Células Endoteliais , Fator VIII , Expressão Gênica , Terapia Genética/métodos , Hemofilia A , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoenxertos , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Modelos Animais de Doenças , Cães , Células Endoteliais/metabolismo , Células Endoteliais/transplante , Fator VIII/biossíntese , Fator VIII/genética , Fator VIII/imunologia , Vetores Genéticos , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/terapia , Humanos , Lentivirus , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Transdução Genética , Transgenes
8.
J Thromb Haemost ; 22(1): 76-89, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37678547

RESUMO

BACKGROUND: One-third of patients with severe hemophilia A develop neutralizing antibodies to the factor VIII (FVIII) protein in response to intravenous replacement therapy. Patients may also generate natural, nonneutralizing antibodies to FVIII before FVIII exposure. These patients are at increased risk of developing neutralizing antibodies to FVIII. However, natural anti-FVIII antibodies are also present in healthy human donors. OBJECTIVES: To further characterize the natural antihuman (h) FVIII antibody repertoire in mice and humans. METHODS: An in-house ELISA was developed using a purified polyclonal immunoglobulin (Ig) standard to quantify anti-hFVIII Ig in cell culture supernatant or plasma from mice (wild-type and FVIII-/-) and adult human donors. RESULTS: All naïve wild-type and FVIII-/- mice, as well as healthy human donors, possess natural anti-hFVIII antibodies. Mice only have natural anti-hFVIII IgM, which is present in germ-free mice, suggesting that they are germline encoded. Although murine marginal zone B cells (MZBs) contribute 44% to all circulating natural IgM, they contribute disproportionately to the anti-hFVIII IgM repertoire (82%). This naturally occurring murine MZB-derived IgM is not B-domain specific and is reduced by intravenously administered hFVIII, suggesting that it may form immune complexes immediately upon hFVIII administration. Natural anti-hFVIII antibodies of IgG, IgM, and IgA isotypes can be detected in adult human donors. There were increased levels of B-domain-favoring anti-hFVIII IgG in 14% of healthy donors, which were markedly different from the rest of the "low-titer" population. CONCLUSIONS: There is a preponderance of natural anti-hFVIII antibodies in both mice and healthy adult human donors.


Assuntos
Fator VIII , Hemofilia A , Adulto , Humanos , Camundongos , Animais , Fator VIII/metabolismo , Imunoglobulina G , Anticorpos Neutralizantes , Imunoglobulina M
9.
Mol Ther ; 19(4): 723-30, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21285959

RESUMO

The objective to use gene therapy to provide sustained, therapeutic levels of factor VIII (FVIII) for hemophilia A is compromised by the emergence of inhibitory antibodies that prevent FVIII from performing its essential function as a cofactor for factor IX (FIX). FVIII appears to be more immunogenic than FIX and an immune response is associated more frequently with FVIII than FIX gene therapy strategies. We have evaluated a modified lentiviral delivery strategy that facilitates liver-restricted transgene expression and prevents off-target expression in hematopoietic cells by incorporating microRNA (miRNA) target sequences. In contrast to outcomes using this strategy to deliver FIX, this modified delivery strategy was in and of itself insufficient to prevent an anti-FVIII immune response in treated hemophilia A mice. However, pseudotyping the lentivirus with the GP64 envelope glycoprotein, in conjunction with a liver-restricted promoter and a miRNA-regulated FVIII transgene resulted in sustained, therapeutic levels of FVIII. These modifications to the lentiviral delivery system effectively restricted FVIII transgene expression to the liver. Plasma levels of FVIII could be increased to around 9% that of normal levels when macrophages were depleted prior to treating the hemophilia A mice with the modified lentiviral FVIII delivery system.


Assuntos
Fator VIII/metabolismo , Hemofilia A/terapia , Lentivirus/genética , MicroRNAs/genética , Animais , Modelos Animais de Doenças , Fator VIII/genética , Terapia Genética/métodos , Camundongos , Camundongos Endogâmicos C57BL
10.
J Thromb Haemost ; 20(11): 2507-2518, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35950488

RESUMO

BACKGROUND: Substantial phenotypic heterogeneity exists in endothelial cells and while much of this heterogeneity results from local microenvironments, epigenetic modifications also contribute. METHODS: Cultured human umbilical vein endothelial cells, human pulmonary microvascular endothelial cells, human hepatic sinusoidal endothelial cells, human lymphatic endothelial cells (hLECs), and two different isolations of endothelial colony forming cells (ECFCs) were assessed for levels of factor VIII (FVIII) and von Willebrand factor (VWF) RNA and protein. The intracellular location and co-localization of both proteins was evaluated with immunofluorescence microscopy and stimulated release toof FVIII and VWF from Weibel-Palade bodies (WPBs) was evaluated. Changes in expression of FVIII and VWF RNA after hLECs and ECFCs were exposed to 2 or 15 dynes/cm2 of laminar shear stress were also assessed. RESULTS: We observed considerable heterogeneity in FVIII and VWF expression among the endothelial cells. With the exception of hLECs, FVIII RNA and protein were barely detectable in any of the endothelial cells and a reciprocal relationship between levels of FVIII and VWF appears to exist. When FVIII and VWF are co-expressed, they do not consistently co-localize in the cytoplasm. However, in hLECs where significantly higher levels of FVIII are expressed, FVIII and VWF co-localize in WPBs and are released together when stimulated. Expression of both FVIII and VWF is markedly reduced when hLECs are exposed to higher or lower levels of laminar shear stress, while in ECFCs there is a minimal response for both proteins. CONCLUSIONS: Variable levels of FVIII and VWF RNA and protein exist in a subset of cultured human endothelial cells. Higher levels of FVIII present in hLECs co-localize with VWF and are released together when exposed to a secretagogue.


Assuntos
Hemostáticos , Fator de von Willebrand , Humanos , Fator de von Willebrand/metabolismo , Fator VIII/genética , Fator VIII/metabolismo , Secretagogos , Células Endoteliais da Veia Umbilical Humana/metabolismo , RNA
11.
Blood ; 113(1): 193-203, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18815284

RESUMO

Non-Fc-receptor binding anti-CD3 Ab therapy, in the setting of several different autoimmune disorders, can induce antigen-specific and long-lasting immunologic tolerance. Because factor VIII (FVIII) inhibitor formation is the most serious treatment-related complication for hemophilia A patients, we tested the efficacy of anti-CD3 to prevent FVIII inhibitor formation in hemophilia A BALB/c and C57BL/6 mice. A short course of low-dose anti-CD3 significantly increased expression of CD25 and the proportion of CD4+CD25+ regulatory T cells in the spleen and potently prevented the production of inhibitory and non-neutralizing anti-FVIII antibodies in both strains of mouse. Depleting the CD4+CD25+ cells during anti-CD3 therapy completely ablated tolerance to FVIII. Further phenotypic characterization of regulatory cells in tolerant mice showed a consistently higher number of CD4+GITR+ and CD4+FoxP3+ cells in both strains of mice. In addition, in tolerant C57BL/6 mice we observed an increase in CD4+CD25+ CTLA-4+ and CD4+CD25+mTGF-beta1+ cells. Finally, in vitro cytokine profiling demonstrated that splenocytes from tolerant BALB/c and C57BL/6 were polarized toward a Th1-immune response. Taken together, these findings indicate that anti-CD3 induces tolerance to FVIII and that the mechanism(s) regulating this response almost certainly occurs through the generation of several distinct regulatory T-cell lineages and by influencing cytokine production and profile.


Assuntos
Anticorpos/farmacologia , Complexo CD3/imunologia , Fator VIII/imunologia , Fator VIII/farmacologia , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Células Th1/imunologia , Animais , Antígenos CD4/metabolismo , Linhagem da Célula/imunologia , Fator VIII/antagonistas & inibidores , Feminino , Hemofilia A/genética , Tolerância Imunológica/imunologia , Imunofenotipagem , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Baço/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo
12.
Blood ; 114(4): 871-80, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19411636

RESUMO

The use of plasma-derived factor VIII (pdFVIII) concentrates in hemophilia A has been reported to result in reduced anti-FVIII antibody formation. In this study, we have investigated whether the cytokine microenvironment induced by pdFVIII has an influence on reducing anti-FVIII antibody titers in hemophilic mice. Microarray and confirmatory quantitative reverse transcription polymerase chain reaction (RT-PCR) experiments show that pdFVIII infusion causes a different transcriptional profile in dendritic cells than recombinant FVIII (rFVIII). Both treatments caused up-regulation of proinflammatory gene expression, but rFVIII and pdFVIII treatments promote expression of genes that induce Th1 and Th2 responses, respectively. Moreover, administration of rFVIII or pdFVIII concentrates resulted in distinct T-cell splenic cytokine microenvironments. rFVIII induced the release of Th1 cytokines and IL-10, whereas pdFVIII induced the release of Th2 cytokines and transforming growth factor-beta. We have also observed high titers of anti-human von Willebrand factor (VWF) antibodies in the pdFVIII-treated mice and propose that this results from antigenic competition. We further investigated the role of this phenomenon using infusions of FVIII and increasing concentrations of recombinant human factor IX (FIX). These studies show an inverse relationship between increasing concentrations of FIX and the production of anti-FVIII antibodies. In summary, these studies report new mechanisms that contribute to reduced anti-FVIII antibody development in hemophilia A after pdFVIII infusions.


Assuntos
Citocinas/metabolismo , Fator VIII/farmacologia , Hemofilia A/metabolismo , Baço/efeitos dos fármacos , Algoritmos , Animais , Formação de Anticorpos/efeitos dos fármacos , Antígeno CD11c/metabolismo , Citocinas/genética , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Fator VIII/imunologia , Fator VIII/isolamento & purificação , Fator VIII/uso terapêutico , Feminino , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemofilia A/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Plasma/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Baço/imunologia , Baço/metabolismo , Baço/patologia , Regulação para Cima/efeitos dos fármacos
13.
Blood ; 114(3): 677-85, 2009 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-19458355

RESUMO

Under certain instances, factor VIII (FVIII) stimulates an immune response, and the resulting neutralizing antibodies present a significant clinical challenge. Immunotherapies to re-establish or induce long-term tolerance would be beneficial, and an in-depth knowledge of mechanisms involved in tolerance induction is essential to develop immune-modulating strategies. We have developed a murine model system for studying mechanisms involved in induction of immunologic tolerance to FVIII in hemophilia A mice. We used lentiviral vectors to deliver the canine FVIII transgene to neonatal hemophilic mice and demonstrated that induction of long-term FVIII tolerance could be achieved. Hemophilia A mice are capable of mounting a robust immune response to FVIII after neonatal gene transfer, and tolerance induction is dependent on the route of delivery and type of promoter used. High-level expression of FVIII was not required for tolerance induction and, indeed, tolerance developed in some animals without evidence of detectable plasma FVIII. Tolerance to FVIII could be adoptively transferred to naive hemophilia recipient mice, and FVIII-stimulated splenocytes isolated from tolerized mice expressed increased levels of interleukin-10 and decreased levels of interleukin-6 and interferon-gamma. Finally, induction of FVIII tolerance mediated by this protocol is associated with a FVIII-expandable population of CD4(+)CD25(+)Foxp3(+) regulatory T cells.


Assuntos
Transferência Adotiva/métodos , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica , Linfócitos T Reguladores/imunologia , Animais , Animais Recém-Nascidos , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Fator VIII/administração & dosagem , Técnicas de Transferência de Genes , Vetores Genéticos , Hemofilia A/terapia , Camundongos , Modelos Animais , Baço/citologia , Linfócitos T/transplante
14.
Blood Adv ; 4(12): 2644-2655, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32556285

RESUMO

The development of neutralizing FVIII antibodies is the most serious complication of hemophilia A treatment. The currently known patient- and treatment-related risk factors for inhibitor development do not accurately predict this adverse event in all patients. The composition of the gut microbiota has been shown to influence immune-mediated diseases at distant anatomical sites (eg, lungs, brain, and joints). We demonstrate that a disrupted gut microbiota can be created in a mouse model of hemophilia A using a broad-spectrum antibiotic. Under controlled conditions, this sustained dysbiosis was associated with an increase in splenic B cells and the development of higher titer, FVIII-specific immunoglobulin G antibodies after FVIII challenge. Splenic and mesenteric lymph node cytokines, T cells, and dendritic cells were unaffected before administration of FVIII. However, the immune transcriptome of both aforementioned secondary lymphoid organs was significantly modified. Short-chain fatty acids (SCFAs), which are immunomodulatory microbial metabolites, were depleted in cecal contents of the dysbiotic mice. Furthermore, supplementation of the drinking water with butyrate, the most immunologically active SCFA, successfully achieved attenuation of the FVIII immune response. Collectively, data from this exploratory study suggest that the composition of the gut microbiota alters the FVIII immune response via the action of specific microbial metabolites on the immune cell transcriptome and that oral supplementation with butyrate effectively reduces the FVIII immune response.


Assuntos
Microbioma Gastrointestinal , Hemofilia A , Animais , Disbiose , Fator VIII , Hemofilia A/tratamento farmacológico , Humanos , Imunidade , Camundongos , Camundongos Endogâmicos C57BL
15.
Front Immunol ; 11: 138, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117285

RESUMO

The development of neutralizing antibodies (inhibitors) against factor VIII (FVIII) is a major complication of hemophilia A treatment. The sole clinical therapy to restore FVIII tolerance in patients with inhibitors remains immune tolerance induction (ITI) which is expensive, difficult to administer and not always successful. Although not fully understood, the mechanism of ITI is thought to rely on inhibition of FVIII-specific B cells (1). Its efficacy might therefore be improved through more aggressive B cell suppression. FcγRIIB is an inhibitory Fc receptor that down-regulates B cell signaling when cross-linked with the B cell receptor (BCR). We sought to investigate if recombinant FVIII Fc (rFVIIIFc), an Fc fusion molecule composed of FVIII and the Fc region of immunoglobulin G1 (IgG1) (2), is able to inhibit B cell activation more readily than FVIII. rFVIIIFc was able to bind FVIII-exposed and naïve B cells from hemophilia A mice as well as a FVIII-specific murine B cell hybridoma line (413 cells). An anti-FcγRIIB antibody and FVIII inhibited binding, suggesting that rFVIIIFc is able to interact with both FcγRIIB and the BCR. Furthermore, incubation of B cells from FVIII-exposed mice and 413 cells with rFVIIIFc resulted in increased phosphorylation of SH-2 containing inositol 5-phosphatase (SHIP) when compared to FVIII. B cells from FVIII-exposed hemophilia A mice also exhibited decreased extracellular signal-regulated kinase (ERK) phosphorylation when exposed to rFVIIIFc. These differences were absent in B cells from naïve, non-FVIII exposed hemophilic mice suggesting an antigen-dependent effect. Finally, rFVIIIFc was able to inhibit B cell calcium flux induced by anti-Ig F(ab)2. Our results therefore indicate that rFVIIIFc is able to crosslink FcγRIIB and the BCR of FVIII-specific B cells, causing inhibitory signaling in these cells.


Assuntos
Linfócitos B/imunologia , Fator VIII/imunologia , Ativação Linfocitária/imunologia , Receptores Fc/imunologia , Receptores de IgG/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linfócitos B/efeitos dos fármacos , Hemofilia A , Fragmentos Fc das Imunoglobulinas/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Receptores de IgG/imunologia , Proteínas Recombinantes de Fusão/imunologia
16.
Res Pract Thromb Haemost ; 3(2): 254-260, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31011709

RESUMO

BACKGROUND: The multimeric glycoprotein von Willebrand factor (VWF) mediates platelet adhesion and aggregation at the site of vessel injury. The adhesive activity of VWF is influenced by its multimer length which is regulated by the metalloprotease ADAMTS13. The ability of ADAMTS13 to regulate platelet thrombus growth in a shear-dependent manner has been described, however, the mechanistic basis of this action has not been well characterized. METHODS: We developed an mCherry-tagged murine ADAMTS13 protein and utilized an ex vivo flow chamber system to visualize the localization of ADAMTS13 within the platelet thrombus under different conditions of shear. Using this system, we also assessed the influence of platelet-mediated tensile force on ADAMTS13 localization within the thrombus using gain-of-function GPIb binding and loss-of-function GPIIbIIIa binding mutants in VWF/ADAMTS13 DKO mice. RESULTS: ADAMTS13 was visualized on the growing platelet thrombus under very high shear using ADAMTS13-mcherry. ADAMTS13-mCherry localized particularly at the top portion of the thrombus and reduced thrombus size as it grew to occlusion. At the pathological high shear of 7500 s-1, platelet-mediated tensile force, involving GPIb but not GPIIbIIIa receptors, influenced localization of ADAMTS13 to the thrombus under conditions of shear. CONCLUSIONS: Tensile force applied on VWF produced by shear stress and platelet GPIb binding has a crucial role in ADAMTS13 activity at the site of thrombus formation. These results suggest that ADAMTS13 activity at the site of platelet thrombus formation is regulated by a shear stress and platelet-dependent feedback mechanism to prevent vessel occlusion and pathological thrombosis.

17.
Stem Cells ; 25(10): 2660-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17615271

RESUMO

Novel therapeutic strategies for hemophilia must be at least as effective as current treatments and demonstrate long-term safety. To date, several small clinical trials of hemophilia gene transfer have failed to show the promise of preclinical evaluations. Therefore, we wanted to develop and evaluate the feasibility of a novel ex vivo gene transfer strategy whereby cells derived from progenitor cells are engineered to express factor VIII (FVIII) and then implanted subcutaneously to act as a depot for FVIII expression. Circulating blood outgrowth endothelial cells (BOECs) were isolated from canine and murine blood and transduced with a lentiviral vector encoding the canine FVIII transgene. To enhance safety, these cells were implanted subcutaneously in a Matrigel scaffold, and the efficacy of this strategy was compared with i.v. delivery of engineered BOECs in nonhemophilic nonobese diabetic/severe combined immunodeficiency mice. Therapeutic levels of FVIII persisted for 15 weeks, and these levels of stable expression were extended to 20 weeks when the cytomegalovirus promoter was replaced with the thrombomodulin regulatory element. Subsequent studies in immunocompetent hemophilic mice, pretreated with tolerizing doses of FVIII or with transient immunosuppression, showed therapeutic FVIII expression for 27 weeks before the eventual return to baseline levels. This loss of transgene expression appears to be due to the disappearance of the implanted cells. The animals treated with either of the two tolerizing regimens did not develop anti-FVIII antibodies. Biodistribution analysis demonstrated that BOECs were retained inside the subcutaneous implants. These results indicate, for the first time, that genetically modified endothelial progenitor cells implanted in a subcutaneous scaffold can provide sustained therapeutic levels of FVIII and are a promising and safe treatment modality for hemophilia A. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células Endoteliais/transplante , Fator VIII/genética , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Hemofilia A/terapia , Lentivirus/genética , Animais , Células Cultivadas/metabolismo , Células Cultivadas/transplante , Dessensibilização Imunológica , Cães , Células Endoteliais/metabolismo , Fator VIII/administração & dosagem , Fator VIII/biossíntese , Fator VIII/imunologia , Estudos de Viabilidade , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Sobrevivência de Enxerto , Hemofilia A/sangue , Hemofilia A/genética , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Alicerces Teciduais , Transdução Genética
18.
Mol Ther Methods Clin Dev ; 2: 15033, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26636112

RESUMO

Recombinant FVIII manufacturing is characterized by poor product stability and low yields. Codon-optimization of transgenes accelerates translation by exploiting the synonymous codon usage bias of a species. However, this can alter the performance of the final product. Additionally, the effects of transgene design across diverse cell types are not well understood and are of interest for next-generation protein and gene therapies. To investigate the effects of transgene design across different host cells, B-domain-deleted (BDD) and modified codon-optimized (CO-N6) transgenes were inserted via lentiviral delivery into cBOECs, HEK293T, and MDCK cells. The CO-N6 cFVIII transgene produced threefold more protein per transgene in HEK293T cells, and sixfold more protein in the two canine cell lines. However, pharmacokinetic analysis in hemophilia A dogs demonstrated that cFVIII produced from cBOECs transduced with the CO-N6 transgene had significantly reduced in vivo recovery. Furthermore, this product showed reduced in vitro stability and activity on thrombin activation versus the BDD product. This trend was reversed in HEK293T lines. Overall, our results demonstrate the need for an integrated approach that not only assesses protein expression levels but also considers the influence that host-cells have on preserving the molecular and biochemical properties of the naturally occurring FVIII.

19.
Hum Gene Ther ; 13(13): 1583-93, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12228013

RESUMO

If gene therapy is to be an effective treatment modality for hemophilia A, therapeutic levels and tissue-restricted expression of factor VIII (FVIII) must be achieved through optimization of transgene expression. To this end, we incorporated three types of sequence elements into a canine B domain-deleted FVIII transgene cassette and individually evaluated their effect on FVIII transgene expression. Functional FVIII activity was initially assessed in vitro and hydrodynamic injection of the different transgene constructs into mice was subsequently used as a model to compare in vivo expression of the various modified transgenes. Our results demonstrate that in vitro transgene expression is, in these studies, not a good predictor of in vivo transgene performance. In vivo analysis of a hybrid tissue-restricted promoter element, consisting of a concatemer of five hepatocyte nuclear factor 1 (HNF-1) consensus-binding motifs juxtaposed to the human FVIII proximal promoter, indicates that it is as efficient at mediating expression of the FVIII protein as the cytomegalovirus promoter. Addition of the full-length canine FVIII 3'-UTR also enhances transgene expression of FVIII in vivo. Sequence analysis of the canine FVIII 3'-UTR and human FVIII 3'-UTR indicates that the former lacks instability sequences and may therefore be more effective in stabilizing FVIII mRNA. Subsequent inclusion of FVIII introns 16 and 17 into the natural locations of the transgene disrupted mRNA processing and abolished expression of the FVIII protein. Introduction of intron 17 proximal to the FVIII cDNA did not enhance in vivo expression of canine FVIII from the transgene.


Assuntos
Regiões 3' não Traduzidas/genética , Fator VIII/genética , Terapia Genética , Hemofilia A/terapia , Fatores de Transcrição/metabolismo , Animais , Cães , Fator VIII/metabolismo , Hemofilia A/genética , Íntrons , Fatores de Transcrição/genética , Transgenes
20.
Thromb Haemost ; 87(4): 659-65, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12008949

RESUMO

We have identified the causative mutation in the hemophilia A dog colony at Queen's University, Canada and have observed a striking similarity with the intron 22 inversion found in approximately 45% of severely affected hemophilia A patients. The canine hemophilia A phenotype arises from aberrant splicing and premature termination of transcription of the FVIII gene, resulting in a polyadenylated transcript lacking exons distal to 22 and terminating with a novel sequence element (NSE). In dogs and other species including humans, this NSE is present in low copy number. One copy of these sequences in the canine genome is within intron 22 and reveals differences in the hybridization banding patterns between normal and hemophilic DNA, suggestive of a large genomic rearrangement. The mutation mechanism may not be uncommon, as identical mutant transcripts were isolated from two hemophilia A littermates that are unrelated to the Queen's colony and from hemophiliac dogs in the colony at Chapel Hill.


Assuntos
Inversão Cromossômica , Doenças do Cão/genética , Cães/genética , Fator VIII/genética , Hemofilia A/veterinária , Splicing de RNA , Animais , Sequência Consenso , Análise Mutacional de DNA , DNA Complementar/genética , Éxons/genética , Biblioteca Gênica , Hemofilia A/genética , Humanos , Íntrons/genética , Masculino , Poliadenilação , Polimorfismo Genético , Sítios de Splice de RNA/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa