Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
1.
J Thromb Haemost ; 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39097231

RÉSUMÉ

Adeno-associated viral vector (AAV) gene therapy provides a promising platform for treatment of monogenic inherited disorders. Clinical studies have demonstrated long-term expression with reduction in bleeding using this approach for the treatment of hemophilia. Despite these advances, there are unknowns surrounding the natural history of recombinant AAV (rAAV) vectors and the cellular mechanisms mediating vector persistence. These unknowns underpin questions regarding long-term efficacy and safety. The predominant mechanism via which AAV is proposed to persist is in circular double-stranded extrachromosomal DNA structures (episomes) within the nucleus. Studies of wild-type (WT-AAV) and rAAV have demonstrated that AAV also persists via integration into a host cell's DNA. It is important to determine whether these integration events can mediate expression or could result in any long-term safety concerns. WT-AAV infection affects a large proportion of the general population, which is thought to have no long-term sequelae. Recent studies have highlighted that this WT-AAV has been detected in cases of acute hepatitis in children and in a minority of cases of hepatocellular carcinoma. Integration following treatment using rAAV has also been reported in preclinical and clinical studies. There have been variable reports on the potential implications of integration for rAAV vectors with data in some murine studies demonstrating recurrent integration with development of hepatocellular carcinoma. These findings have not been seen in other pre-clinical or clinical studies. In this review, we will summarize current understanding of the natural history of AAV (wild-type and recombinant) with a focus on genomic integration and the cellular implications.

2.
Blood ; 143(23): 2373-2385, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38452208

RÉSUMÉ

ABSTRACT: Gene therapy using adeno-associated virus (AAV) vectors is a promising approach for the treatment of monogenic disorders. Long-term multiyear transgene expression has been demonstrated in animal models and clinical studies. Nevertheless, uncertainties remain concerning the nature of AAV vector persistence and whether there is a potential for genotoxicity. Here, we describe the mechanisms of AAV vector persistence in the liver of a severe hemophilia A dog model (male = 4, hemizygous; and female = 4, homozygous), more than a decade after portal vein delivery. The predominant vector form was nonintegrated episomal structures with levels correlating with long-term transgene expression. Random integration was seen in all samples (median frequency, 9.3e-4 sites per cell), with small numbers of nonrandom common integration sites associated with open chromatin. No full-length integrated vectors were found, supporting predominant episomal vector-mediated long-term transgene expression. Despite integration, this was not associated with oncogene upregulation or histopathological evidence of tumorigenesis. These findings support the long-term safety of this therapeutic modality.


Sujet(s)
Dependovirus , Facteur VIII , Thérapie génétique , Vecteurs génétiques , Hémophilie A , Foie , Animaux , Chiens , Dependovirus/génétique , Hémophilie A/génétique , Hémophilie A/thérapie , Vecteurs génétiques/génétique , Foie/métabolisme , Foie/anatomopathologie , Mâle , Thérapie génétique/méthodes , Femelle , Facteur VIII/génétique , Techniques de transfert de gènes , Intégration virale , Transgènes , Modèles animaux de maladie humaine
3.
Sci Transl Med ; 16(735): eadh0027, 2024 Feb 21.
Article de Anglais | MEDLINE | ID: mdl-38381848

RÉSUMÉ

Antifibrinolytic drugs are used extensively for on-demand treatment of severe acute bleeding. Controlling fibrinolysis may also be an effective strategy to prevent or lessen chronic recurring bleeding in bleeding disorders such as hemophilia A (HA), but current antifibrinolytics have unfavorable pharmacokinetic profiles. Here, we developed a long-lasting antifibrinolytic using small interfering RNA (siRNA) targeting plasminogen packaged in clinically used lipid nanoparticles (LNPs) and tested it to determine whether reducing plasmin activity in animal models of HA could decrease bleeding frequency and severity. Treatment with the siRNA-carrying LNPs reduced circulating plasminogen and suppressed fibrinolysis in wild-type and HA mice and dogs. In HA mice, hemostatic efficacy depended on the injury model; plasminogen knockdown improved hemostasis after a saphenous vein injury but not tail vein transection injury, suggesting that saphenous vein injury is a murine bleeding model sensitive to the contribution of fibrinolysis. In dogs with HA, LNPs carrying siRNA targeting plasminogen were as effective at stabilizing clots as tranexamic acid, a clinical antifibrinolytic, and in a pilot study of two dogs with HA, the incidence of spontaneous or excess bleeding was reduced during 4 months of prolonged knockdown. Collectively, these data demonstrate that long-acting antifibrinolytic therapy can be achieved and that it provides hemostatic benefit in animal models of HA.


Sujet(s)
Antifibrinolytiques , Hémophilie A , Hémostatiques , Liposomes , Nanoparticules , Chiens , Animaux , Souris , Fibrinolyse/génétique , Antifibrinolytiques/pharmacologie , Plasminogène/pharmacologie , Hémophilie A/traitement médicamenteux , Petit ARN interférent , Projets pilotes , Hémorragie/traitement médicamenteux , Hémostatiques/pharmacologie
4.
Mol Cell ; 83(17): 3049-3063.e6, 2023 09 07.
Article de Anglais | MEDLINE | ID: mdl-37591243

RÉSUMÉ

Cohesin connects CTCF-binding sites and other genomic loci in cis to form chromatin loops and replicated DNA molecules in trans to mediate sister chromatid cohesion. Whether cohesin uses distinct or related mechanisms to perform these functions is unknown. Here, we describe a cohesin hinge mutant that can extrude DNA into loops but is unable to mediate cohesion in human cells. Our results suggest that the latter defect arises during cohesion establishment. The observation that cohesin's cohesion and loop extrusion activities can be partially separated indicates that cohesin uses distinct mechanisms to perform these two functions. Unexpectedly, the same hinge mutant can also not be stopped by CTCF boundaries as well as wild-type cohesin. This suggests that cohesion establishment and cohesin's interaction with CTCF boundaries depend on related mechanisms and raises the possibility that both require transient hinge opening to entrap DNA inside the cohesin ring.


Sujet(s)
Protéines du cycle cellulaire , Chromatides , Humains , Chromatides/génétique , Sites de fixation , Protéines du cycle cellulaire/génétique , Protéines chromosomiques nonhistones/génétique ,
5.
EMBO J ; 42(16): e113475, 2023 08 15.
Article de Anglais | MEDLINE | ID: mdl-37357575

RÉSUMÉ

Genetic information is stored in linear DNA molecules, which are highly folded inside cells. DNA replication along the folded template path yields two sister chromatids that initially occupy the same nuclear region in an intertwined arrangement. Dividing cells must disentangle and condense the sister chromatids into separate bodies such that a microtubule-based spindle can move them to opposite poles. While the spindle-mediated transport of sister chromatids has been studied in detail, the chromosome-intrinsic mechanics presegregating sister chromatids have remained elusive. Here, we show that human sister chromatids resolve extensively already during interphase, in a process dependent on the loop-extruding activity of cohesin, but not that of condensins. Increasing cohesin's looping capability increases sister DNA resolution in interphase nuclei to an extent normally seen only during mitosis, despite the presence of abundant arm cohesion. That cohesin can resolve sister chromatids so extensively in the absence of mitosis-specific activities indicates that DNA loop extrusion is a generic mechanism for segregating replicated genomes, shared across different Structural Maintenance of Chromosomes (SMC) protein complexes in all kingdoms of life.


Sujet(s)
Chromatides , Protéines chromosomiques nonhistones , Humains , Chromatides/génétique , Chromatides/métabolisme , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/métabolisme , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Mitose , ADN , Phase G2 ,
6.
Nature ; 609(7925): 183-190, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35922507

RÉSUMÉ

Dividing eukaryotic cells package extremely long chromosomal DNA molecules into discrete bodies to enable microtubule-mediated transport of one genome copy to each of the newly forming daughter cells1-3. Assembly of mitotic chromosomes involves DNA looping by condensin4-8 and chromatin compaction by global histone deacetylation9-13. Although condensin confers mechanical resistance to spindle pulling forces14-16, it is not known how histone deacetylation affects material properties and, as a consequence, segregation mechanics of mitotic chromosomes. Here we show how global histone deacetylation at the onset of mitosis induces a chromatin-intrinsic phase transition that endows chromosomes with the physical characteristics necessary for their precise movement during cell division. Deacetylation-mediated compaction of chromatin forms a structure dense in negative charge and allows mitotic chromosomes to resist perforation by microtubules as they are pushed to the metaphase plate. By contrast, hyperacetylated mitotic chromosomes lack a defined surface boundary, are frequently perforated by microtubules and are prone to missegregation. Our study highlights the different contributions of DNA loop formation and chromatin phase separation to genome segregation in dividing cells.


Sujet(s)
Chromatine , Microtubules , Mitose , Acétylation , Chromatine/métabolisme , Ségrégation des chromosomes , ADN/métabolisme , Histone/métabolisme , Microtubules/métabolisme , Transition de phase , Appareil du fuseau/métabolisme
7.
Res Pract Thromb Haemost ; 6(5): e12747, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35814801

RÉSUMÉ

The ISTH London 2022 Congress is the first held (mostly) face-to-face again since the COVID-19 pandemic took the world by surprise in 2020. For 2 years we met virtually, but this year's in-person format will allow the ever-so-important and quintessential creativity and networking to flow again. What a pleasure and joy to be able to see everyone! Importantly, all conference proceedings are also streamed (and available recorded) online for those unable to travel on this occasion. This ensures no one misses out. The 2022 scientific program highlights new developments in hemophilia and its treatment, acquired and other inherited bleeding disorders, thromboinflammation, platelets and coagulation, clot structure and composition, fibrinolysis, vascular biology, venous thromboembolism, women's health, arterial thrombosis, pediatrics, COVID-related thrombosis, vaccine-induced thrombocytopenia with thrombosis, and omics and diagnostics. These areas are elegantly reviewed in this Illustrated Review article. The Illustrated Review is a highlight of the ISTH Congress. The format lends itself very well to explaining the science, and the collection of beautiful graphical summaries of recent developments in the field are stunning and self-explanatory. This clever and effective way to communicate research is revolutionary and different from traditional formats. We hope you enjoy this article and will be inspired by its content to generate new research ideas.

8.
Mol Ther Methods Clin Dev ; 26: 61-71, 2022 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-35782594

RÉSUMÉ

Recombinant adeno-associated virus (AAV) is an effective platform for therapeutic gene transfer; however, tissue-tropism differences between species are a challenge for successful translation of preclinical results to humans. We evaluated the use of in vitro primary hepatocyte cultures to predict in vivo liver-directed AAV expression in different species. We assessed whether in vitro AAV transduction assays in cultured primary hepatocytes from mice, nonhuman primates (NHPs), and humans could model in vivo liver-directed AAV expression of valoctocogene roxaparvovec (AAV5-hFVIII-SQ), an experimental gene therapy for hemophilia A with a hepatocyte-selective promoter. Relative levels of DNA and RNA in hepatocytes grown in vitro correlated with in vivo liver transduction across species. Expression in NHP hepatocytes more closely reflected expression in human hepatocytes than in mouse hepatocytes. We used this hepatocyte culture model to assess transduction efficacy of a novel liver-directed AAV capsid across species and identified which of 3 different canine factor VIII vectors produced the most transgene expression. Results were confirmed in vivo. Further, we determined mechanisms mediating inhibition of AAV5-hFVIII-SQ expression by concomitant isotretinoin using primary human hepatocytes. These studies support using in vitro primary hepatocyte models to predict species translatability of liver-directed AAV gene therapy and improve mechanistic understanding of drug-drug interactions.

9.
Int J Lab Hematol ; 43 Suppl 1: 117-123, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34288447

RÉSUMÉ

Since the cloning and characterization of the factor VIII (FVIII) and factor IX genes in the mid-1980s, gene therapy has been perceived as having significant potential for the treatment of severe hemophilia. Now, some 35 years later, these proposals are close to being realized through the licensing of the first clinical gene therapy product. Adeno-associated viral vector-mediated gene therapy for hemophilia A and B has been extensively investigated in preclinical models over the past 20 years, and since 2011, there has been increasing evidence in early phase clinical trials that this therapeutic strategy can provide safe and effective rescue of the hemostatic phenotype in severe hemophilia. As the uptake of hemophilia gene therapy progresses, it is clear that many aspects of the gene therapy process require crucial laboratory support to ensure safe and effective outcomes from his new therapeutic paradigm. These laboratory contributions extend from evaluations of the gene therapy vehicle, assessments of the patient immune status for the vector, and ultimately the performance of assays to determine the hemostatic benefit of the gene therapy and potentially of its long-term safety on the host genome. As with many aspects of past hemophilia care, the safe and effective delivery of gene therapy will require an informed and coordinated contribution from laboratory science.


Sujet(s)
Thérapie génétique , Hémophilie A/thérapie , Hémophilie B/thérapie , Animaux , Coagulation sanguine , Tests de coagulation sanguine , Essais cliniques comme sujet , Dependovirus/génétique , Facteur IX/génétique , Facteur VIII/génétique , Thérapie génétique/effets indésirables , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/effets indésirables , Vecteurs génétiques/génétique , Hémophilie A/sang , Hémophilie A/diagnostic , Hémophilie A/génétique , Hémophilie B/sang , Hémophilie B/diagnostic , Hémophilie B/génétique , Humains , Mutation , Phénotype , Résultat thérapeutique
10.
Haemophilia ; 27(3): 490-499, 2021 May.
Article de Anglais | MEDLINE | ID: mdl-33650732

RÉSUMÉ

INTRODUCTION: Inhibitor formation is the greatest challenge facing persons with haemophilia treated with factor concentrates. The gold standard testing methodologies are the Nijmegen-Bethesda assay (NBA) for FVIII and Bethesda assay (BA) for FIX inhibitors, which are affected by pre-analytical and inter-laboratory variability. AIMS: To evaluate inhibitor testing methodology and assess correlation between self-reported and actual methodology. METHODS: Methodology was evaluated using a survey distributed alongside a UK National External Quality Assessment Service Blood Coagulation external quality assurance (EQA) exercise for FVIII and FIX inhibitor testing. RESULTS: Seventy four survey and EQA exercise responses were received (response rate 63.2%), with 50 paired survey/EQA results. 47.1% (33/70) reported using the NBA and 42.9% (30/70) the BA for FVIII inhibitor testing. Review of FVIII inhibitor assay methodology demonstrated discrepancy (self-reported to actual) in 64.3% (BA reporting) and 27.6% (NBA reporting). Pre-analytical heat treatment was used by 32.4%, most commonly 56°C for 30 minutes. Assay cut-offs of 0.1-1.0 BU/mL were reported. EQA samples (acquired FVIII and congenital FIX) demonstrated titres and coefficients of variation (CV) of 3.1 BU/mL (0.7-15.4 BU/mL; CV = 43%) and 18.0 BU/mL (0-117 BU/mL; CV = 33%), respectively. No significant assay or laboratory factors were found to explain this variance, which could have resulted in change in management for 6 patients (5 misclassified high-titre FVIII inhibitors and 1 false negative for a FIX inhibitor). CONCLUSIONS: Heterogeneity was seen at each stage of assay methodology. No assay-related factors were found to explain variation in inhibitor titres. Further standardization is required to improve inhibitor quantification to guide patient care.


Sujet(s)
Facteur VIII , Hémophilie A , Inhibiteurs des facteurs de la coagulation sanguine , Tests de coagulation sanguine , Hémophilie A/diagnostic , Hémophilie A/traitement médicamenteux , Humains , Royaume-Uni
11.
Hemasphere ; 5(3): e540, 2021 Mar.
Article de Anglais | MEDLINE | ID: mdl-33604517

RÉSUMÉ

The clinical potential of hemophilia gene therapy has now been pursued for the past 30 years, and there is a realistic expectation that this goal will be achieved within the next couple of years with the licensing of a gene therapy product. While recent late phase clinical trials of hemophilia gene therapy have shown promising results, there remain a number of issues that require further attention with regard to both efficacy and safety of this therapeutic approach. In this review, we present information relating to the current status of the field and focus attention on the unanswered questions for hemophilia gene therapy and the future challenges that need to be overcome to enable the widespread application of this treatment paradigm.

12.
Blood Coagul Fibrinolysis ; 31(7): 452-458, 2020 Oct.
Article de Anglais | MEDLINE | ID: mdl-32833805

RÉSUMÉ

: Women with inherited bleeding disorders (IBDs) are reported to have higher rates of primary and secondary postpartum haemorrhage (PPH), even with optimal haemostatic management. We evaluated whether women with IBD have higher odds of PPH compared with those without, when controlled for mode of delivery with a control group of women without IBDs. The obstetric experiences and outcomes of all women with IBD delivering at a tertiary centre between 2008 and 2017, were compared with matched controls (1 : 1). Obstetric care was provided according to national guidelines to both women with IBD and controls. Primary PPH was defined as estimated blood loss at least 500 ml. There were 46 completed pregnancies in women with IBD: 16 haemophilia A carriers, eight haemophilia B carriers, eight factor XI deficiency patients and 14 von Willebrand disease patients (type 1 = 6; type 2 = 8). No peripartum haemostatic treatment was received by carriers of haemophilia A or B. There were 74 control pregnancies. Women with IBD had higher odds of primary PPH, in a model controlling for mode of anaesthesia (adjusted odds ratio 5.30, 95% confidence interval 1.02-27.59, P = 0.048). Carriers of haemophilia A had a higher, statistically nonsignificant, odds for primary PPH than controls (adjusted odds ratio 6.85, confidence interval 0.77-60.73, P = 0.084). An increase in primary PPH was observed in women with IBD, particularly in haemophilia A, despite management according to guidelines. These results warrant further investigation and consideration should be given as to which factor levels to target.


Sujet(s)
Troubles héréditaires de la coagulation sanguine/complications , Hémorragie de la délivrance/diagnostic , Adulte , Études de cohortes , Femelle , Humains , Jeune adulte
13.
Br J Haematol ; 189(1): 39-53, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32064603

RÉSUMÉ

Anti-drug antibody formation following factor VIII (FVIII) replacement therapy is the most important treatment-related complication in patients with severe haemophilia A. A significant number of these antibodies show neutralising activity against FVIII and are referred to as FVIII inhibitors. Alloimmunity to FVIII, given the absence of endogenous circulating FVIII protein, may be predictable to some extent; however, only 30% of patients develop inhibitors. Genetic and environmental risk factors have been identified, contributing to the likelihood of inhibitor development. Multiple immunological theories have been proposed which in part explain the outcomes of many epidemiological studies. Significant differences exist among replacement therapies, including the source, FVIII sequence, glycosylation, formulation components, impurities and aggregation potential, which significantly complicate interpretation of the results from these studies. In this review, we present recent advances in the understanding of the cellular mechanisms of inhibitor formation and highlight some areas of uncertainty requiring further investigation.


Sujet(s)
Autoanticorps , Inhibiteurs des facteurs de la coagulation sanguine , Facteur VIII , Autoanticorps/génétique , Autoanticorps/immunologie , Inhibiteurs des facteurs de la coagulation sanguine/génétique , Inhibiteurs des facteurs de la coagulation sanguine/immunologie , Facteur VIII/génétique , Facteur VIII/immunologie , Facteur VIII/usage thérapeutique , Hémophilie A/traitement médicamenteux , Hémophilie A/génétique , Hémophilie A/immunologie , Hémophilie A/anatomopathologie , Humains , Mâle , Facteurs de risque
14.
Hum Mol Genet ; 28(R1): R95-R101, 2019 10 01.
Article de Anglais | MEDLINE | ID: mdl-31332444

RÉSUMÉ

Hemophilia is an X-linked inherited bleeding disorder, resulting from defects in the F8 (hemophilia A) or F9 (hemophilia B) genes. Persons with hemophilia have bleeding episodes into the soft tissues and joints, which are treated with self-infusion of factor VIII or IX concentrates. Hemophilia provides an attractive target for gene therapy studies, due to the monogenic nature of these disorders and easily measurable endpoints (factor levels and bleed rates). All successful, pre-clinical and clinical studies to date have utilized recombinant adeno-associated viral (AAV) vectors for factor VIII or IX hepatocyte transduction. Recent clinical data have presented normalization of factor levels in some patients with improvements in bleed rate and quality of life. The main toxicity seen within these studies has been early transient elevation in liver enzymes, with variable effect on transgene expression. Although long-term data are awaited, durable expression has been seen within the hemophilia dog model with no late-toxicity or oncogenesis. There are a number of phase III studies currently recruiting; however, there may be some limitations in translating these data to clinical practice, due to inclusion/exclusion criteria. AAV-based gene therapy is one of a number of novel approaches for treatment of hemophilia with other gene therapy (in vivo and ex vivo) and non-replacement therapies progressing through clinical trials. Availability of these high-cost novel therapeutics will require evolution of both clinical and financial healthcare services to allow equitable personalization of care for persons with hemophilia.


Sujet(s)
Facteur IX/génétique , Facteur VIII/génétique , Thérapie génétique , Hémophilie A/génétique , Hémophilie A/thérapie , Hémophilie B/génétique , Hémophilie B/thérapie , Animaux , Études cliniques comme sujet , Association thérapeutique , Régulation de l'expression des gènes , Thérapie génétique/effets indésirables , Thérapie génétique/méthodes , Vecteurs génétiques/effets indésirables , Vecteurs génétiques/génétique , Humains , Alloanticorps/immunologie , Spécificité d'organe , Transduction génétique , Transgènes , Résultat thérapeutique
15.
Trends Cell Biol ; 29(9): 717-726, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31230958

RÉSUMÉ

During mitosis, replicated chromosomes segregate such that each daughter cell receives one copy of the genome. Faithful mechanical transport during mitosis requires that chromosomes undergo extensive structural changes as the cell cycle progresses, resulting in the formation of compact, cylindrical bodies. Such structural changes encompass a range of different activities, including longitudinal condensation of the chromosome axis, global chromatin compaction, resolution of sister chromatids, and individualisation of chromosomes into separate bodies. After mitosis, chromosomes undergo further reorganisation to rebuild interphase cell nuclei. Here we review the requirements for mitotic chromosomes to successfully transmit genetic information to daughter cells and the biophysical principles that underpin such requirements.


Sujet(s)
Protéines du cycle cellulaire/génétique , Chromatine/métabolisme , Chromosomes/génétique , Chromosomes/ultrastructure , Animaux , Protéines du cycle cellulaire/métabolisme , Chromatides/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Altération de l'ADN , Réparation de l'ADN , Réplication de l'ADN , Protéines fongiques/métabolisme , Génome , Humains , Mitose , Complexes multiprotéiques/métabolisme , Protéines nucléaires/métabolisme
18.
Br J Haematol ; 176(5): 796-804, 2017 Mar.
Article de Anglais | MEDLINE | ID: mdl-28198996

RÉSUMÉ

Inhibitor formation in non-severe haemophilia A is a life-long risk and associated with morbidity and mortality. There is a paucity of data to understand real-world inhibitor screening practice. We evaluated the treatment burden, haemostatic strategies, F8 genotyping and inhibitor screening practices in non-severe haemophilia A in seven London haemophilia centres. In the 2-year study period, 44% (377/853) patients received at least one haemostatic treatment. Seventy-nine percent of those treated (296/377) received factor VIII (FVIII) concentrate. F8 genotype was known in 88% (331/377) of individuals. Eighteen per cent (58/331) had 'high-risk' F8 genotypes. In patients with 'standard-risk' F8 genotypes treated on-demand with FVIII concentrate, 51·3% episodes (243/474) were screened within 1 year. However, poor screening compliance was observed after 'high-risk' treatment episodes. In patients with 'standard-risk' F8 genotypes, 12·3% (28/227) of treatment episodes were screened in the subsequent 6 weeks after surgery or a bleed requiring ≥5 exposure days. Similarly, in the context of 'high-risk' F8 genotypes after any FVIII exposure, only 13·6% (12/88) of episodes were screened within 6 weeks. Further study is required to assess optimal practice of inhibitor screening in non-severe haemophilia A to inform subsequent clinical decisions and provide more robust prevalence data to further understand the underlying immunological mechanism.


Sujet(s)
Facteur VIII/génétique , Génotype , Hémophilie A/immunologie , Hémophilie A/thérapie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Enfant , Enfant d'âge préscolaire , Hémophilie A/génétique , Hémostatiques/usage thérapeutique , Humains , Nourrisson , Dépistage de masse/méthodes , Adulte d'âge moyen , Études rétrospectives , Appréciation des risques , Jeune adulte
19.
Mol Cell ; 61(4): 563-574, 2016 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-26895425

RÉSUMÉ

Sister chromatid cohesion conferred by entrapment of sister DNAs within a tripartite ring formed between cohesin's Scc1, Smc1, and Smc3 subunits is created during S and destroyed at anaphase through Scc1 cleavage by separase. Cohesin's association with chromosomes is controlled by opposing activities: loading by Scc2/4 complex and release by a separase-independent releasing activity as well as by cleavage. Coentrapment of sister DNAs at replication is accompanied by acetylation of Smc3 by Eco1, which blocks releasing activity and ensures that sisters remain connected. Because fusion of Smc3 to Scc1 prevents release and bypasses the requirement for Eco1, we suggested that release is mediated by disengagement of the Smc3/Scc1 interface. We show that mutations capable of bypassing Eco1 in Smc1, Smc3, Scc1, Wapl, Pds5, and Scc3 subunits reduce dissociation of N-terminal cleavage fragments of Scc1 (NScc1) from Smc3. This process involves interaction between Smc ATPase heads and is inhibited by Smc3 acetylation.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Protéines de Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Acétylation , Sites de fixation , Protéines du cycle cellulaire/composition chimique , Protéines du cycle cellulaire/génétique , Protéines chromosomiques nonhistones/composition chimique , Protéines chromosomiques nonhistones/génétique , ADN fongique/métabolisme , Modèles moléculaires , Mutation , Structure tertiaire des protéines , Protéines de Saccharomyces cerevisiae/composition chimique , Protéines de Saccharomyces cerevisiae/génétique ,
20.
Thromb Haemost ; 114(4): 804-11, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26063073

RÉSUMÉ

Antibody formation to factor VIII (FVIII) remains the greatest clinical and diagnostic challenge to the haemophilia-treating physician. Current guidance for testing for inhibitory FVIII antibodies (inhibitors) recommends the functional Nijmegen-Bethesda assay (NBA). A FVIII ELISA offers a complementary, immunological approach for FVIII antibody testing. It was the aim of this study to retrospectively evaluate the performance of a FVIII ELISA (index) for detection of FVIII antibodies, compared with the NBA (reference). All samples sent for routine FVIII antibody testing at two haemophilia Comprehensive Care Centres, were tested in parallel using the NBA and a solid-phase, indirect FVIII ELISA kit (Immucor). A total of 497 samples from 239 patients (severe haemophilia A=140, non-severe haemophilia A=85, acquired haemophilia A=14) were available for analysis. Sixty-three samples tested positive by the NBA (prevalence 12.7%, 95% confidence interval [CI], 9.9-15.9 %), with a median inhibitor titre of 1.2 BU/ml (range 0.7-978.0). The FVIII ELISA demonstrated a specificity of 94.0% (95%CI, 91.3-96.0), sensitivity of 77.8% (95%CI, 65.5-87.3), negative predictive value of 96.7% (95%CI, 94.5-98.2), positive predictive value 65.3% (95%CI, 53.5-76.0), negative likelihood ratio 0.2 (95%CI, 0.1-0.4), positive likelihood ratio 13.0 (95%CI, 8.7-19.3) and a diagnostic odds ratio of 54.9 (95%CI, 27.0-112.0). Strong positive correlation (r=0.77, p<0.001) was seen between the results of the NBA (log adjusted) and FVIII ELISA optical density. In conclusion, FVIII ELISA offers a simple, specific, surveillance method enabling batch testing of non-urgent samples for the presence of FVIII antibodies.


Sujet(s)
Autoanticorps/sang , Coagulants/immunologie , Test ELISA , Facteur VIII/immunologie , Hémophilie A/diagnostic , Analyse de variance , Marqueurs biologiques/sang , Coagulants/usage thérapeutique , Facteur VIII/usage thérapeutique , Hémophilie A/sang , Hémophilie A/traitement médicamenteux , Hémophilie A/génétique , Hémophilie A/immunologie , Humains , Fonctions de vraisemblance , Londres , Biais de l'observateur , Odds ratio , Valeur prédictive des tests , Trousses de réactifs pour diagnostic , Reproductibilité des résultats , Études rétrospectives , Indice de gravité de la maladie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE