Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.872
Filtrer
1.
J Cell Mol Med ; 28(11): e18460, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38864710

RÉSUMÉ

Haemophilic arthropathy (HA), a common comorbidity in haemophilic patients leads to joint pain, deformity and reduced quality of life. We have recently demonstrated that a long non-coding RNA, Neat1 as a primary regulator of matrix metalloproteinase (MMP) 3 and MMP13 activity, and its induction in the target joint has a deteriorating effect on articular cartilage. In the present study, we administered an Adeno-associated virus (AAV) 5 vector carrying an short hairpin (sh)RNA to Neat1 via intra-articular injection alone or in conjunction with systemic administration of a capsid-modified AAV8 (K31Q) vector carrying F8 gene (F8-BDD-V3) to study its impact on HA. AAV8K31Q-F8 vector administration at low dose, led to an increase in FVIII activity (16%-28%) in treated mice. We further observed a significant knockdown of Neat1 (~40 fold vs. untreated injured joint, p = 0.005) in joint tissue of treated mice and a downregulation of chondrodegenerative enzymes, MMP3, MMP13 and the inflammatory mediator- cPLA2, in mice receiving combination therapy. These data demonstrate that AAV mediated Neat1 knockdown in combination with F8 gene augmentation can potentially impact mediators of haemophilic joint disease.


Sujet(s)
Dependovirus , Facteur VIII , Vecteurs génétiques , Hémophilie A , Matrix Metalloproteinase 13 , Matrix metalloproteinase 3 , ARN long non codant , Animaux , Hémophilie A/génétique , Hémophilie A/thérapie , Hémophilie A/complications , Dependovirus/génétique , ARN long non codant/génétique , Matrix Metalloproteinase 13/métabolisme , Matrix Metalloproteinase 13/génétique , Souris , Matrix metalloproteinase 3/génétique , Matrix metalloproteinase 3/métabolisme , Vecteurs génétiques/génétique , Vecteurs génétiques/administration et posologie , Facteur VIII/génétique , Facteur VIII/métabolisme , Maladies articulaires/thérapie , Maladies articulaires/génétique , Maladies articulaires/étiologie , Humains , Thérapie génétique/méthodes , Souris de lignée C57BL , Cartilage articulaire/métabolisme , Cartilage articulaire/anatomopathologie , Modèles animaux de maladie humaine , Mâle
2.
Blood Coagul Fibrinolysis ; 35(5): 238-247, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38874909

RÉSUMÉ

The aim of this study is to characterize zebrafish coagulation cofactors fviii and fv mutant fish and assess if they phenocopy classical hemophilia A and factor V deficiency in humans. The embryos from fviii and fv zebrafish heterozygote mutants generated by ENU mutagenesis were purchased from the ZIRC repository. They were reared to adulthood and genotyped. The heterozygote male and female were crossed to get homozygote, heterozygote, and wild-type fish. Functional kinetic coagulation assays and bleeding assays were performed on normal and mutant adult fish, and venous laser injury assays were performed on the larvae. The DNA from fviii and fv mutants were sequenced to confirm if they have a premature stop codon in exon 19, and in exon 2, respectively, and in both mutants, the amino acid glutamine is replaced with a stop codon. Homozygous and heterozygous 5 days post fertilization (dpf) larvae for fviii and fv deficient mutants exhibited prolonged time to occlusion after venous laser injury compared to wild-type controls. The homozygous and heterozygous fviii adult mutants showed modest bleeding and delayed fibrin formation in the kinetic partial thromboplastin time (kPTT) assay with their plasma. fv homozygous larvae had poor survival beyond 12 dpf. However, heterozygous fv mutants exhibited heavy bleeding and prolonged fibrin formation in the kPTT and kPT assay compared with wild-type siblings. Our characterization showed fviii and fv mutants from ZIRC phenocopied to a considerable extent classical hemophilia A and factor V deficiency in humans, respectively. These models should be useful in studying and developing novel drugs that reverse the phenotype and in generating suppressor mutations to identify novel factors that compensate for these deficiencies.


Sujet(s)
Modèles animaux de maladie humaine , Déficit en facteur V , Facteur VIII , Hémophilie A , Danio zébré , Animaux , Hémophilie A/génétique , Hémophilie A/sang , Facteur VIII/génétique , Facteur VIII/métabolisme , Déficit en facteur V/génétique , Proaccélérine/génétique , Mutation , Femelle , Mâle , Coagulation sanguine , Humains
3.
Thromb Res ; 240: 109061, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870771

RÉSUMÉ

BACKGROUND: Von Willebrand Disease (VWD) is the most common inherited bleeding disorder. VWD is characterized by an abnormal quantity or quality of von Willebrand Factor (VWF). Anemia is often found at presentation for a bleeding disorder evaluation due to chronic blood loss. OBJECTIVES/HYPOTHESIS: We hypothesized that anemia is associated with elevations in both VWF and factor VIII (FVIII) over baseline. We also hypothesized that obesity would be associated with increased levels of VWF. METHODS: We conducted a single-center review of the electronic health record for patients that had proximal von Willebrand profiles and Hb data. RESULTS: We identified 4552 unique subjects with VWF studies and a CBC within 24 h. We found that decreasing hemoglobin inversely correlated with VWF antigen, VWF ristocetin cofactor activity, and FVIII activity. We also found that obesity and Black race were independently associated with increased VWF antigen, activity, and FVIII activity. Hb, race, and body mass index (BMI) continued to be determinants of VWF and FVIII levels in multivariable analysis. CONCLUSION: Our study demonstrates that anemia, race, and BMI were found to be associated with elevation of VWF antigen, VWF activity, and FVIII levels. As many individuals with anemia present for evaluation for a bleeding disorder, these variables need to be considered. KEY POINTS: - Anemia was found to be associated with elevation of VWF antigen, VWF activity and FVIII levels. - Testing von Willebrand factor at times of anemia may mask a diagnosis of von Willebrand Disease.


Sujet(s)
Indice de masse corporelle , Facteur VIII , Hémoglobines , Facteur de von Willebrand , Humains , Facteur de von Willebrand/analyse , Facteur de von Willebrand/métabolisme , Facteur VIII/analyse , Facteur VIII/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Adulte , Hémoglobines/analyse , Maladies de von Willebrand/sang , Anémie/sang , Sujet âgé , Obésité/sang , Obésité/complications
4.
J Thromb Haemost ; 22(8): 2171-2183, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38718928

RÉSUMÉ

BACKGROUND: Replacement and nonreplacement therapies effectively control bleeding in hemophilia A (HA) but imply lifelong interventions. Authorized gene addition therapy could provide a cure but still poses questions on durability. FVIIIgene correction would definitively restore factor (F)VIII production, as shown in animal models through nuclease-mediated homologous recombination (HR). However, low efficiency and potential off-target double-strand break still limit HR translatability. OBJECTIVES: To correct common model single point mutations leading to severe HA through the recently developed double-strand break/HR-independent base editing (BE) and prime editing (PE) approaches. METHODS: Screening for efficacy of BE/PE systems in HEK293T cells transiently expressing FVIII variants and validation at DNA (sequencing) and protein (enzyme-linked immunosorbent assay; activated partial thromboplastin time) level in stable clones. Evaluation of rescue in engineered blood outgrowth endothelial cells by lentiviral-mediated delivery of BE. RESULTS: Transient assays identified the best-performing BE/PE systems for each variant, with the highest rescue of FVIII expression (up to 25% of wild-type recombinant FVIII) for the p.R2166∗ and p.R2228Q mutations. In stable clones, we demonstrated that the mutation reversion on DNA (∼24%) was consistent with the rescue of FVIII secretion and activity of 20% to 30%. The lentiviral-mediated delivery of the selected BE systems was attempted in engineered blood outgrowth endothelial cells harboring the p.R2166∗ and p.R2228Q variants, which led to an appreciable and dose-dependent rescue of secreted functional FVIII. CONCLUSION: Overall data provide the first proof-of-concept for effective BE/PE-mediated correction of HA-causing mutations, which encourage studies in mouse models to develop a personalized cure for large cohorts of patients through a single intervention.


Sujet(s)
Facteur VIII , Édition de gène , Thérapie génétique , Hémophilie A , Facteur VIII/génétique , Facteur VIII/métabolisme , Humains , Hémophilie A/génétique , Hémophilie A/thérapie , Hémophilie A/sang , Édition de gène/méthodes , Cellules HEK293 , Cellules endothéliales/métabolisme , Mutation ponctuelle , Mutation , Systèmes CRISPR-Cas
5.
Acta Anaesthesiol Scand ; 68(7): 940-948, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38723274

RÉSUMÉ

BACKGROUND: Understanding the recovery of post-COVID-19 organ dysfunction is essential. We evaluated coagulation 6 months post-COVID-19, examining its recovery and association with lung function. METHODS: Patients treated for COVID-19 at intensive care units between 3/2020 and 1/2021 were analyzed for complete blood count (CBC) and coagulation biomarkers (prothrombin time activity (%) (PT%), activated partial thromboplastin time (APTT), fibrinogen, coagulation factor VIII (FVIII), antithrombin (AT), and D-dimer) during the 6 months post-hospitalization. Results were compared with acute phase values and correlated with pulmonary function tests (PFT), including forced vital capacity (FVC) and hemoglobin-corrected diffusing capacity percentage of predicted (DLCOc%), recorded 6 months post-hospitalization. We examined the association between coagulation biomarkers and DLCOc% using linear regression with age, sex, and invasive mechanical ventilation (IMV) duration, and FVIII (correlated with DLCOc%) as covariates. RESULTS: Most CBCs and coagulation biomarkers had median values within the normal range. However, only 21% (15/70) of patients achieved full normalization of all biomarkers. Compared to acute COVID-19, hemoglobin, PT%, and AT increased, while leukocytes, fibrinogen, FVIII, and D-dimer decreased. Despite decreased levels, FVIII remained elevated in 46% (31/68), leukocytes in 26% (18/70), and D-dimer in 27% (18/67) at 6 months. A weak negative correlation (r = -0.37, p = .036) was found between DLCOc% and FVIII. Multivariable analysis revealed a weak, independent association between DLCOc% and FVIII. Excluding patients with anticoagulation therapy, FVIII no longer correlated with DLCOc%, while AT showed a moderate correlation with DLCOc%. CONCLUSION: Only a few patients had normal CBC and coagulation biomarker values 6 months after critical COVID-19. A weak negative correlation between DLCOc% and FVIII suggests that deranged coagulation activity may be associated with reduced diffusing capacity.


Sujet(s)
Marqueurs biologiques , COVID-19 , Humains , COVID-19/sang , Mâle , Femelle , Adulte d'âge moyen , Marqueurs biologiques/sang , Sujet âgé , Hémogramme , Tests de la fonction respiratoire , Coagulation sanguine/physiologie , Produits de dégradation de la fibrine et du fibrinogène/analyse , Adulte , Poumon/physiopathologie , Tests de coagulation sanguine , Fibrinogène/analyse , Fibrinogène/métabolisme , Ventilation artificielle , Facteur VIII/analyse , Facteur VIII/métabolisme
6.
Blood Adv ; 8(15): 3929-3940, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38820442

RÉSUMÉ

ABSTRACT: A2 domain dissociation in activated factor VIII (FVIIIa) results in reduced activity. Previous studies demonstrated that some FVIII mutants (D519V/E665V and K1813A) with delayed A2 dissociation enhanced coagulation potential. We speculated, therefore, that FVIII encompassing a combination of these mutations might further enhance coagulant activity. The aim was to assess the D519V/E665V/K1813A-FVIII mutation as a gain of function. The FVIII mutants, D519V/E665V/K1813A, D519V/E665V, and K1813A were expressed in a baby hamster kidney cell system, and global coagulation potential of these mutants was compared with wild-type (WT) FVIII in vitro and in hemophilia A mice in vivo. Kinetic analyses indicated that the apparent Kd for FIXa on the tenase assembly with D519V/E665V and D519V/E665V/K1813A mutants were lower, and that the generated FXa for D519V/E665V/K1813A was significantly greater than WT-FVIII. WT-FVIII activity after thrombin activation increased by ∼12-fold within 5 minutes, and returned to initial levels within 30 minutes. In contrast, The FVIII-related activity of D519V/E665V/K1813A increased further with time after thrombin activation, and showed an ∼25-fold increase at 2 hours. The A2 dissociation rate of D519V/E665V/K1813A was ∼50-fold slower than the WT in a 1-stage clotting assay. Thrombin generation assays demonstrated that D519V/E665V/K1813A (0.125 nM) exhibited coagulation potential comparable with that of the WT (1 nM). In animal studies, rotational thromboelastometry and tail-clip assays showed that the coagulation potential of D519V/E665V/K1813A (0.25 µg/kg) was equal to that of the WT (2 µg/kg). FVIII-D519V/E665V/K1813A mutant could provide an approximately eightfold increase in hemostatic function of WT-FVIII because of increased FVIIIa stability and the association between FVIIIa and FIXa.


Sujet(s)
Coagulation sanguine , Facteur VIII , Hémophilie A , Mutation , Animaux , Facteur VIII/génétique , Facteur VIII/métabolisme , Souris , Humains , Hémophilie A/génétique , Hémophilie A/sang , Cricetinae , Thrombine/métabolisme , Substitution d'acide aminé , Lignée cellulaire , Modèles animaux de maladie humaine
9.
Expert Rev Hematol ; 17(7): 341-351, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38752747

RÉSUMÉ

INTRODUCTION: Hereditary thrombotic thrombocytopenic purpura (hTTP) is caused by deficiency of plasma ADAMTS13 activity, resulting from ADAMTS13 mutations. ADAMTS13 cleaves ultra large von Willebrand factor (VWF), thus reducing its multimer sizes. Hereditary deficiency of plasma ADAMTS13 activity leads to the formation of excessive platelet-VWF aggregates in small arterioles and capillaries, resulting in hTTP. AREAS COVERED: PubMed search from 1956 to 2024 using thrombotic thrombocytopenic purpura and therapy identified 3,675 articles. Only the articles relevant to the topic were selected for discussion, which focuses on pathophysiology, clinical presentations, and mechanisms of action of emerging therapeutics for hTTP. Current therapies include infusion of plasma, or coagulation factor VIII, or recombinant ADAMTS13. Emerging therapies include anti-VWF A1 aptamers or nanobody and gene therapies with adeno-associated viral vector or self-inactivated lentiviral vector or a sleeping beauty transposon system for a long-term expression of a functional ADAMTS13 enzyme. EXPERT OPINION: Frequent plasma infusion remains to be the standard of care in most parts of the world, while recombinant ADAMTS13 has become the treatment of choice for hTTP in some of the Western countries. The success of gene therapies in preclinical models may hold a promise for future development of these novel approaches for a cure of hTTP.


Sujet(s)
Protéine ADAMTS13 , Thérapie génétique , Purpura thrombotique thrombocytopénique , Humains , Purpura thrombotique thrombocytopénique/thérapie , Purpura thrombotique thrombocytopénique/génétique , Protéine ADAMTS13/génétique , Protéine ADAMTS13/métabolisme , Protéine ADAMTS13/déficit , Facteur de von Willebrand/métabolisme , Facteur de von Willebrand/génétique , Anticorps à domaine unique/usage thérapeutique , Prise en charge de la maladie , Facteur VIII/génétique , Facteur VIII/usage thérapeutique , Facteur VIII/métabolisme , Mutation , Animaux
10.
EMBO Mol Med ; 16(6): 1427-1450, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38684862

RÉSUMÉ

Lentiviral vectors (LV) are efficient vehicles for in vivo gene delivery to the liver. LV integration into the chromatin of target cells ensures their transmission upon proliferation, thus allowing potentially life-long gene therapy following a single administration, even to young individuals. The glycoprotein of the vesicular stomatitis virus (VSV.G) is widely used to pseudotype LV, as it confers broad tropism and high stability. The baculovirus-derived GP64 envelope protein has been proposed as an alternative for in vivo liver-directed gene therapy. Here, we perform a detailed comparison of VSV.G- and GP64-pseudotyped LV in vitro and in vivo. We report that VSV.G-LV transduced hepatocytes better than GP64-LV, however the latter showed improved transduction of liver sinusoidal endothelial cells (LSEC). Combining GP64-pseudotyping with the high surface content of the phagocytosis inhibitor CD47 further enhanced LSEC transduction. Coagulation factor VIII (FVIII), the gene mutated in hemophilia A, is naturally expressed by LSEC, thus we exploited GP64-LV to deliver a FVIII transgene under the control of the endogenous FVIII promoter and achieved therapeutic amounts of FVIII and correction of hemophilia A mice.


Sujet(s)
Cellules endothéliales , Facteur VIII , Thérapie génétique , Vecteurs génétiques , Hémophilie A , Lentivirus , Foie , Animaux , Hémophilie A/thérapie , Hémophilie A/génétique , Vecteurs génétiques/génétique , Cellules endothéliales/métabolisme , Souris , Lentivirus/génétique , Thérapie génétique/méthodes , Foie/métabolisme , Facteur VIII/génétique , Facteur VIII/métabolisme , Humains , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/métabolisme , Transduction génétique/méthodes , Hépatocytes/métabolisme , Hépatocytes/virologie , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme
11.
Am J Hypertens ; 37(8): 580-587, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38642910

RÉSUMÉ

BACKGROUND: Nearly half of all Americans have hypertension, and Black adults experience a disproportionate burden. Hypercoagulability may relate to hypertension risk, and higher levels of factor VIII increase thrombosis risk. Black adults have higher factor VIII and more hypertension than other groups. Whether higher factor VIII associates with incident hypertension is unknown. METHODS: The Biomarkers as Mediators of Racial Disparities in Risk Factors (BioMedioR) study measured certain biomarkers in a sex-race stratified sample of 4,400 REGARDS participants who attended both visits. We included BioMedioR participants, excluding those with prevalent hypertension, missing factor VIII level, or covariates of interest. Modified Poisson regression estimated risk ratios (RR) for incident hypertension by higher log-transformed factor VIII level per SD (SD of log-transformed factor VIII, 0.33). Weighting was applied to take advantage of REGARDS sampling design. RESULTS: Among the 1,814 participants included (55% female, 24% Black race), the median follow-up was 9.5 years and 35% (2,146/6,138) developed hypertension. Black participants had a higher median (IQR) factor VIII level (105.6%; 87.1%-126.9%) than White participants (95.6%; 79.8%-115.9%; P < 0.001). The age- and sex-adjusted Black-White hypertension RR was 1.45 (95% CI 1.28, 1.63). Higher factor VIII was not associated with more hypertension (final model RR 1.01; 95% CI 0.94, 1.07). CONCLUSIONS: In a prospective study of Black and White adults without prevalent hypertension, factor VIII was not associated with greater hypertension risk.


Sujet(s)
Facteur VIII , Disparités de l'état de santé , Hypertension artérielle , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Marqueurs biologiques/sang , , Pression sanguine , Facteur VIII/analyse , Facteur VIII/métabolisme , Hypertension artérielle/ethnologie , Hypertension artérielle/épidémiologie , Hypertension artérielle/physiopathologie , Incidence , Études prospectives , Appréciation des risques , Facteurs de risque , Accident vasculaire cérébral/ethnologie , Accident vasculaire cérébral/épidémiologie , Accident vasculaire cérébral/sang , États-Unis/épidémiologie , Blanc
12.
J Thromb Haemost ; 22(7): 1894-1908, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38679335

RÉSUMÉ

BACKGROUND: von Willebrand disease (VWD) is the most common inherited bleeding disorder, characterized by either partial or complete von Willebrand factor (VWF) deficiency or by the occurrence of VWF proteoforms of altered functionality. The gene encoding VWF is highly polymorphic, giving rise to a variety of proteoforms with varying plasma concentrations and clinical significance. OBJECTIVES: To address this complexity, we translated genomic variation in VWF to corresponding VWF proteoforms circulating in blood. METHODS: VWF was characterized in VWD patients (n = 64) participating in the Willebrand in the Netherlands study by conventional laboratory testing, DNA sequencing and complementary discovery, and targeted mass spectrometry-based plasma proteomic strategies. RESULTS: Unbiased plasma profiling combined with immune enrichment of VWF verified VWF and its binding partner factor VIII as key determinants of VWD and revealed a remarkable heterogeneity in VWF amino acid sequence coverage among patients. Subsequent VWF proteotyping enabled identification of both polymorphisms (eg, p.Thr789Ala, p.Gln852Arg, and p.Thr1381Ala), as well as pathogenic variants (n = 16) along with their corresponding canonical sequences. Targeted proteomics using stable isotope-labeled peptides confirmed unbiased proteotyping for 5 selected variants and suggested differential proteoform quantities in plasma. The variant-to-wild-type peptide ratio was determined in 6 type 2B patients heterozygous for p.Arg1306Trp, confirming the relatively low proteoform concentration of the pathogenic variant. The elevated VWF propeptide/VWF ratio indicated increased clearance of specific VWF proteoforms. CONCLUSION: This study highlights how VWF proteotyping from plasma could be the first step to bridge the gap between genotyping and functional testing in VWD.


Sujet(s)
Protéomique , Maladies de von Willebrand , Facteur de von Willebrand , Humains , Facteur de von Willebrand/génétique , Facteur de von Willebrand/analyse , Facteur de von Willebrand/métabolisme , Maladies de von Willebrand/diagnostic , Maladies de von Willebrand/sang , Maladies de von Willebrand/génétique , Protéomique/méthodes , Pays-Bas , Phénotype , Femelle , Facteur VIII/génétique , Facteur VIII/analyse , Facteur VIII/métabolisme , Spectrométrie de masse , Mâle , Valeur prédictive des tests
13.
Thromb Res ; 237: 184-195, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38631156

RÉSUMÉ

BACKGROUND AND AIMS: Blood disorders, such as sickle cell disease, and other clinical conditions are often accompanied by intravascular hemolytic events along with the development of severe coagulopathies. Hemolysis, in turn, leads to the accumulation of Fe(II/III)-protoporphyrin IX (heme) in the intravascular compartment, which can trigger a variety of proinflammatory and prothrombotic reactions. As such, heme binding to the blood coagulation proteins factor VIII (FVIII), fibrinogen, and activated protein C with functional consequences has been demonstrated earlier. METHODS: We herein present an in-depth characterization of the FVIII-heme interaction at the molecular level and its (patho-)physiological relevance through the application of biochemical, biophysical, structural biology, bioinformatic, and diagnostic tools. RESULTS: FVIII has a great heme-binding capacity with seven heme molecules associating with the protein. The respective binding sites were identified by investigating heme binding to FVIII-derived peptides in combination with molecular docking and dynamic simulation studies of the complex as well as cryo-electron microscopy, revealing three high-affinity and four moderate heme-binding motifs (HBMs). Furthermore, the relevance of the FVIII-heme complex formation was characterized in physiologically relevant assay systems, revealing a ~ 50 % inhibition of the FVIII cofactor activity even in the protein-rich environment of blood plasma. CONCLUSION: Our study provides not only novel molecular insights into the FVIII-heme interaction and its physiological relevance, but also strongly suggests the reduction of the intrinsic pathway and the accentuation of the final clotting step (by, for example, fibrinogen crosslinking) in hemolytic conditions as well as a future perspective in the context of FVIII substitution therapy of hemorrhagic events in hemophilia A patients.


Sujet(s)
Facteur VIII , Hème , Humains , Sites de fixation , Coagulation sanguine , Facteur VIII/métabolisme , Facteur VIII/composition chimique , Hème/métabolisme , Simulation de docking moléculaire , Liaison aux protéines , Relation structure-activité
14.
Eur J Immunol ; 54(4): e2350506, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38429238

RÉSUMÉ

Tolerance to self-proteins involves multiple mechanisms, including conventional CD4+ T-cell (Tconv) deletion in the thymus and the recruitment of natural regulatory T cells (nTregs). The significant incidence of autoantibodies specific for the blood coagulation factor VIII (FVIII) in healthy donors illustrates that tolerance to self-proteins is not always complete. In contrast to FVIII-specific Tconvs, FVIII-specific nTregs have never been revealed and characterized. To determine the frequency of FVIII-specific Tregs in human peripheral blood, we assessed the specificity of in vitro expanded Tregs by the membrane expression of the CD137 activation marker. Amplified Tregs maintain high levels of FOXP3 expression and exhibit almost complete demethylation of the FOXP3 Treg-specific demethylated region. The cells retained FOXP3 expression after long-term culture in vitro, strongly suggesting that FVIII-specific Tregs are derived from the thymus. From eleven healthy donors, we estimated the frequencies of FVIII-specific Tregs at 0.17 cells per million, which is about 10-fold lower than the frequency of FVIII-specific CD4+ T cells we previously published. Our results shed light on the mechanisms of FVIII tolerance by a renewed approach that could be extended to other self- or non-self-antigens.


Sujet(s)
Facteur VIII , Hémophilie A , Humains , Facteur VIII/métabolisme , Lymphocytes T régulateurs , Hémophilie A/métabolisme , Autoanticorps , Facteurs de transcription Forkhead/métabolisme
15.
Mol Ther ; 32(4): 969-981, 2024 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-38341614

RÉSUMÉ

The ability to target the native production site of factor VIII (FVIII)-liver sinusoidal endothelial cells (LSECs)-can improve the outcome of hemophilia A (HA) gene therapy. By testing a matrix of ultrasound-mediated gene delivery (UMGD) parameters for delivering a GFP plasmid into the livers of HA mice, we were able to define specific conditions for targeted gene delivery to different cell types in the liver. Subsequently, two conditions were selected for experiments to treat HA mice via UMGD of an endothelial-specific human FVIII plasmid: low energy (LE; 50 W/cm2, 150 µs pulse duration) to predominantly target endothelial cells or high energy (HE; 110 W/cm2, 150 µs pulse duration) to predominantly target hepatocytes. Both groups of UMGD-treated mice achieved persistent FVIII activity levels of ∼10% over 84 days post treatment; however, half of the HE-treated mice developed low-titer inhibitors while none of the LE mice did. Plasma transaminase levels and histological liver examinations revealed minimal transient liver damage that was lower in the LE group than in the HE group. These results indicate that UMGD can safely target LSECs with a lower-energy condition to achieve persistent FVIII gene expression, demonstrating that this novel technology is highly promising for therapeutic correction of HA.


Sujet(s)
Facteur VIII , Hémophilie A , Humains , Facteur VIII/métabolisme , Hémophilie A/génétique , Hémophilie A/thérapie , Hémophilie A/anatomopathologie , Cellules endothéliales/métabolisme , Hépatocytes/métabolisme , Foie/métabolisme , Thérapie génétique/méthodes
16.
Blood ; 143(18): 1845-1855, 2024 May 02.
Article de Anglais | MEDLINE | ID: mdl-38320121

RÉSUMÉ

ABSTRACT: Coagulation factor VIII (FVIII) and its carrier protein von Willebrand factor (VWF) are critical to coagulation and platelet aggregation. We leveraged whole-genome sequence data from the Trans-Omics for Precision Medicine (TOPMed) program along with TOPMed-based imputation of genotypes in additional samples to identify genetic associations with circulating FVIII and VWF levels in a single-variant meta-analysis, including up to 45 289 participants. Gene-based aggregate tests were implemented in TOPMed. We identified 3 candidate causal genes and tested their functional effect on FVIII release from human liver endothelial cells (HLECs) and VWF release from human umbilical vein endothelial cells. Mendelian randomization was also performed to provide evidence for causal associations of FVIII and VWF with thrombotic outcomes. We identified associations (P < 5 × 10-9) at 7 new loci for FVIII (ST3GAL4, CLEC4M, B3GNT2, ASGR1, F12, KNG1, and TREM1/NCR2) and 1 for VWF (B3GNT2). VWF, ABO, and STAB2 were associated with FVIII and VWF in gene-based analyses. Multiphenotype analysis of FVIII and VWF identified another 3 new loci, including PDIA3. Silencing of B3GNT2 and the previously reported CD36 gene decreased release of FVIII by HLECs, whereas silencing of B3GNT2, CD36, and PDIA3 decreased release of VWF by HVECs. Mendelian randomization supports causal association of higher FVIII and VWF with increased risk of thrombotic outcomes. Seven new loci were identified for FVIII and 1 for VWF, with evidence supporting causal associations of FVIII and VWF with thrombotic outcomes. B3GNT2, CD36, and PDIA3 modulate the release of FVIII and/or VWF in vitro.


Sujet(s)
Molécules d'adhérence cellulaire , Facteur VIII , Kininogènes , Lectines de type C , Récepteurs de surface cellulaire , Facteur de von Willebrand , Humains , Facteur de von Willebrand/génétique , Facteur de von Willebrand/métabolisme , Facteur VIII/génétique , Facteur VIII/métabolisme , Polymorphisme de nucléotide simple , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Analyse de randomisation mendélienne , Étude d'association pangénomique , Thrombose/génétique , Thrombose/sang , Études d'associations génétiques , Mâle , Cellules endothéliales/métabolisme , Femelle
17.
J Thromb Haemost ; 22(5): 1489-1495, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38325597

RÉSUMÉ

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.


Sujet(s)
Plaquettes , Facteur VIII , Activation plaquettaire , Agrégation plaquettaire , Glycoprotéines de membrane plaquettaire , Humains , Glycoprotéines de membrane plaquettaire/métabolisme , Activation plaquettaire/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Phosphorylation , Facteur VIII/métabolisme , Collagène/métabolisme , Liaison aux protéines , Cytométrie en flux , Thrombine/métabolisme , Relation dose-effet des médicaments , Microscopie confocale
18.
J Biotechnol ; 384: 45-54, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38403131

RÉSUMÉ

Recently developed multi-specific antibody formats enable new therapeutic concepts. Conveniently, formats with an Fc domain allow purification in well-established mAb platform processes. However, due to the structural complexity of the formats, the assembled molecules may be sensitive to extreme pH commonly used for viral inactivation. An alternative to low pH incubation for virus inactivation is the use of a mixture of tri-n-butyl phosphate (TnBP, solvent) and Polysorbate 80 (PS80, detergent). While TnBP is toxic, this combination has a long history of use in the manufacturing of human plasma-derived products that are sensitive to low or high pH incubation. Data are provided demonstrating that the solvent/detergent (S/D) treatment using TnBP and PS80 can be successfully used for pH-sensitive, multi-specific antibody formats in the clarified cell culture fluid (CCCF). A different placement of the S/D within the purification process, namely during the capture by Protein A (PA), has been evaluated. This alternative placement allows effective viral inactivation by S/D while preserving the viral reduction and viral inactivation achieved through the PA step itself, enabling the cumulation of these effects. Furthermore, the process alternative simplifies the liquid handling by reducing the added volumes of the required S/D liquids, thus reducing the amount of toxic TnBP to a minimum. Data are shown demonstrating a complete removal of TnBP and PS80 in the process.


Sujet(s)
Détergents , Inactivation virale , Humains , Facteur VIII/métabolisme , Anticorps , Solvants , Concentration en ions d'hydrogène
19.
Gen Thorac Cardiovasc Surg ; 72(8): 512-518, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38280130

RÉSUMÉ

OBJECTIVE: Degenerative aortic stenosis and coronary artery disease are considered to be the most prevalent cardiovascular diseases in industrialized countries. This study aims to determine the change over time in von Willebrand factor antigen, von Willebrand factor activity, and factor VIII and where there is a correlation with total post-operative drainage. METHODS: The single-center retrospective study included 203 consecutive patients (64.5% male), undergoing coronary artery bypass surgery between March 1, 2019 and June 30, 2020 at the University Clinical Center of Serbia in the Clinic for Cardiac Surgery in Belgrade, Serbia. All patients 18 years or older who presented with isolated, hemodynamically significant aortic stenosis were included. The control group consisted of patients who presented with only coronary artery disease. RESULTS: Between patients with only coronary artery disease and patients with coronary artery diseases and aortic stenosis, there was a statistically significant difference between pre-op and 1-month post-op fibrinogen, factor VIII, von Willebrand factor antigen, and von Willebrand factor (p < 0.001), post-op drainage, with overall lower drainage in coronary artery disease patients, and consistent increase in von Willebrand factor antigen, von Willebrand factor activity, and Factor VIII post-operatively in patients with coronary artery diseases and aortic stenosis. CONCLUSION: This study has shown that there is a correlation between von Willebrand factor antigen, von Willebrand factor activity and total drainage to the level of statistical significance in aortic stenosis patients and in the overall study population.


Sujet(s)
Sténose aortique , Pontage aortocoronarien , Maladie des artères coronaires , Facteur VIII , Maladies de von Willebrand , Facteur de von Willebrand , Humains , Mâle , Sténose aortique/chirurgie , Sténose aortique/complications , Femelle , Maladies de von Willebrand/sang , Maladies de von Willebrand/complications , Maladies de von Willebrand/épidémiologie , Études rétrospectives , Maladie des artères coronaires/chirurgie , Maladie des artères coronaires/complications , Sujet âgé , Facteur de von Willebrand/analyse , Facteur de von Willebrand/métabolisme , Adulte d'âge moyen , Facteur VIII/analyse , Facteur VIII/métabolisme , Pontage aortocoronarien/effets indésirables , Drainage , Marqueurs biologiques/sang , Résultat thérapeutique , Facteurs temps , Serbie/épidémiologie , Sujet âgé de 80 ans ou plus
20.
J Thromb Haemost ; 22(1): 76-89, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37678547

RÉSUMÉ

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.


Sujet(s)
Facteur VIII , Hémophilie A , Adulte , Humains , Souris , Animaux , Facteur VIII/métabolisme , Immunoglobuline G , Anticorps neutralisants , Immunoglobuline M
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE