51
|
Lombardi AM, Cabrio L, Zanon E, Sartori MT, Navaglia F, Plebani M, Girolami A. A Large Deletion due to a New Mutation (Intron 13/Exon 23) in a Sporadic Case of Severe Hemophilia A. Clin Appl Thromb Hemost 2016; 10:61-4. [PMID: 14979407 DOI: 10.1177/107602960401000110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A case of sporadic hemophilia A in a young child was investigated from a molecular biology point of view. The propositus is a 4-year-old severe hemophiliac who was first seen when he was 2 years old. At that time, easy bruising and hematomas were noted because of accidental falls while toddling. The coagulation study showeda prolonged partial thromboplastin time and a factor VIII level of 1.3% of normal. Molecular biologic analysis showed a large deletion involving intron 13 up to exon 23. In the inversion study, the propositus exhibited only a 10 kb band, and this result suggests that intron 22 was deleted while his mother shows a normal pattern. To further examine the length of the deletion, a long polymerase chain reaction by means of primers amplifying the region from exon 13 to 23. In the index patient, an approximate 13-kb product was obtained, whereas no product was obtained from his mother. The mother investigated by means of polymorphism was shown not to be a carrier.
Collapse
Affiliation(s)
- Anna Maria Lombardi
- Department of Medical and Surgical Sciences, 2nd Chair of Internal Medicine, University of Padua Medical School, Padua, Italy.
| | | | | | | | | | | | | |
Collapse
|
52
|
Abstract
Hemophilia A, a deficiency in the activity of coagulation factor (F) VIII, is an X-linked bleeding disorder with an approximate incidence of one in 5,000 male infants. Bleeding-related complications often result in greater severity of disease, poor quality of life, surgical interventions for severe joint destruction, and shortened life span. With the availability of plasma-derived and recombinant FVIII products, the benefits of primary prophylaxis were demonstrated and is now the standard of care for patients with severe factor deficiencies. Current hemophilia research is focusing on the creation of new factor replacement therapies with longer half-lives; accessing alternative mechanisms to achieve desired hemostasis and enhance bypassing activity; and limiting the immunogenicity of the protein. PEGylation involves the covalent attachment of polyethylene glycol (PEG) to a protein, peptide, or a small molecule drug. PEG effectively increases the molecular weight and size of the protein by creating a hydrophilic cloud around the molecule. This molecular change may reduce susceptibility of the molecule to proteolytic activity and degradation. It is also believed that PEGylation changes the surface charge of the protein that ultimately interferes with some receptor-mediated clearance processes. The half-life of PEGylated factor is more prolonged when compared to non-PEGylated full-length recombinant FVIII. The dawn of a new era in the care of hemophilia patients is upon us with the release of recombinant FVIII products with extended half-lives, and products with even more extended half-life will become available in a very short time. With all the promise of these new agents, many questions still remain.
Collapse
Affiliation(s)
- Tung Thanh Wynn
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Florida, Gainesville, FL
| | - Burak Gumuscu
- Pediatric Hematology-Oncology, Bon Secours Health System, St. Mary's Hospital, Richmond, VA; Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
53
|
Abstract
There are numerous new coagulation factor concentrates that are being manufactured using recombinant technology. Some are available for use currently for the treatment of bleeding disorders and others are in clinical trials. Recombinant factor VIII concentrates are licenced in most countries and one FVIII concentrate with the B domain deleted should be available in the near future. Recombinant VIIa concentrate is in advanced phase III testing for the treatment of patients with inhibitor antibodies. Recombinant factor IX has been used successfully in animals and will be tested in humans shortly. This paper reviews these products and discusses their uses and possible side-effects.
Collapse
Affiliation(s)
- D B Brettler
- New England Hemophilia Center, The Medical Center of Central Massachusetts, 119 Belmont Street, Worcester, MA 01605-2892, USA
| |
Collapse
|
54
|
Monaghan M, Wakabayashi H, Griffiths AE, Fay PJ. Stabilizing interactions between D666-S1787 and T657-Y1792 at the A2-A3 interface support factor VIIIa stability in the blood clotting pathway. J Thromb Haemost 2016; 14:1021-30. [PMID: 26878264 PMCID: PMC4870127 DOI: 10.1111/jth.13292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/20/2016] [Indexed: 12/01/2022]
Abstract
UNLABELLED Essentials Factor VIIIa (FVIIIa) is unstable due to loss of A2; D666 and Y1792 contribute to its stability. We conducted a study to identify the interactions made at these residues at the A2-A3 interface. We present evidence for stabilizing interactions between D666-S1787 and T657-Y1792 in FVIIIa. A D666C/S1788C variant with a disulfide A2-A3 linkage has a FVIIIa decay rate that is 1% of wild-type. SUMMARY Background Factor (F)VIIIa activity and stability depends on the non-covalent association of the A2 subunit with the A1/A3C1C2 dimer, but the interactions that contribute to A2 association are not well defined. Previous work had shown that D666A and Y1792F mutations at the A2-A3 interface resulted in increased FVIIIa decay, suggesting that the residues were involved in bonding interactions important for FVIIIa stability. Objectives Several potential hydrogen bonding partners of D666 and Y1792 across the A2-A3 interface were selected from the low-resolution FVIII crystal structure, and we used mutagenesis and biochemical analysis to examine the bonding interactions occurring at D666 and Y1792. Methods Using a series of stability and functional analyses, we analyzed FVIII variants in which D666 and Y1792 were each swapped with the residues of potential bonding partners. Results and conclusions We present evidence for hydrogen bonds between D666 and S1787 and between Y1792 and T657 that are important for FVIIIa stability. D666S/S1787D and T657Y/Y1792T variants each displayed wild-type (WT)-like FVIIIa stability and performed like WT FVIII in a series of functional analyses, whereas D666S, S1787D, and Y1792T single variants showed increased FVIIIa decay and a T657Y variant had little FVIIIa activity. These results suggest that WT hydrogen bonds are disrupted with the single mutations but maintained in the swap variants. Furthermore, mutation of D666 and S1788 to cysteine resulted in disulfide bond formation across the A2-A3 interface, confirming the close proximity of D666 and S1787, and this covalent attachment of the A2 subunit significantly increased FVIIIa stability.
Collapse
Affiliation(s)
- M Monaghan
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - H Wakabayashi
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - AE Griffiths
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - PJ Fay
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| |
Collapse
|
55
|
Nichols TC, Hough C, Agersø H, Ezban M, Lillicrap D. Canine models of inherited bleeding disorders in the development of coagulation assays, novel protein replacement and gene therapies. J Thromb Haemost 2016; 14:894-905. [PMID: 26924758 DOI: 10.1111/jth.13301] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/08/2016] [Indexed: 01/09/2023]
Abstract
Animal models of inherited bleeding disorders are important for understanding disease pathophysiology and are required for preclinical assessment of safety prior to testing of novel therapeutics in human and veterinary medicine. Experiments in these animals represent important translational research aimed at developing safer and better treatments, such as plasma-derived and recombinant protein replacement therapies, gene therapies and immune tolerance protocols for antidrug inhibitory antibodies. Ideally, testing is done in animals with the analogous human disease to provide essential safety information, estimates of the correct starting dose and dose response (pharmacokinetics) and measures of efficacy (pharmacodynamics) that guide the design of human trials. For nearly seven decades, canine models of hemophilia, von Willebrand disease and other inherited bleeding disorders have not only informed our understanding of the natural history and pathophysiology of these disorders but also guided the development of novel therapeutics for use in humans and dogs. This has been especially important for the development of gene therapy, in which unique toxicities such as insertional mutagenesis, germ line gene transfer and viral toxicities must be assessed. There are several issues regarding comparative medicine in these species that have a bearing on these studies, including immune reactions to xenoproteins, varied metabolism or clearance of wild-type and modified proteins, and unique tissue tropism of viral vectors. This review focuses on the results of studies that have been performed in dogs with inherited bleeding disorders that closely mirror the human condition to develop safe and effective protein and gene-based therapies that benefit both species.
Collapse
Affiliation(s)
- T C Nichols
- Departments of Medicine and Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - C Hough
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - H Agersø
- Research and Development Novo Nordisk A/S, Maaloev, Denmark
| | - M Ezban
- Research and Development Novo Nordisk A/S, Maaloev, Denmark
| | - D Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
56
|
O'Sullivan JM, Jenkins PV, Rawley O, Gegenbauer K, Chion A, Lavin M, Byrne B, O'Kennedy R, Preston RJS, Brophy TM, O'Donnell JS. Galectin-1 and Galectin-3 Constitute Novel-Binding Partners for Factor VIII. Arterioscler Thromb Vasc Biol 2016; 36:855-63. [PMID: 27013611 DOI: 10.1161/atvbaha.115.306915] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 03/14/2016] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Recent studies have demonstrated that galectin-1 (Gal-1) and galectin-3 (Gal-3) can bind von Willebrand factor and directly modulate von Willebrand factor-dependent early thrombus formation in vivo. Because the glycans expressed on human factor VIII (FVIII) are similar to those of von Willebrand factor, we investigated whether galectins might also bind and modulate the activity of FVIII. APPROACH AND RESULTS Immunosorbant assays and surface plasmon resonance analysis confirmed that Gal-1 and Gal-3 bound purified FVIII with high affinity. Exoglycosidase removal of FVIII N-linked glycans significantly reduced binding to both Gal-1 and Gal-3. Moreover, combined removal of both the N- and O-glycans of FVIII further attenuated Gal-3 binding. Notably, specific digestion of FVIII high-mannose glycans at N239 and N2118 significantly impaired FVIII affinity for Gal-1. Importantly Gal-1, but not Gal-3, bound to free FVIII in the plasma milieu, and significantly inhibited FVIII functional activity. Interestingly, commercial recombinant FVIII (rFVIII) concentrates are manufactured in different cell lines and differ in their glycosylation profiles. Although the biological mechanism has not been defined, recent studies in previously untreated patients with severe hemophilia A reported significant differences in inhibitor development associated with different rFVIII products. Interestingly, Gal-1 and Gal-3 both displayed enhanced affinity for BHK-rFVIII compared with CHO-rFVIII. Furthermore, binding of Gal-1 and Gal-3 to BDD-FVIII was markedly reduced compared with full-length rFVIII. CONCLUSIONS We have identified Gal-1 and Gal-3 as novel-binding partners for human FVIII and demonstrated that Gal-1 binding can influence the procoagulant activity of FVIII.
Collapse
Affiliation(s)
- Jamie M O'Sullivan
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - P Vince Jenkins
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Orla Rawley
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Kristina Gegenbauer
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Alain Chion
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Michelle Lavin
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Barry Byrne
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Richard O'Kennedy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Roger J S Preston
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - Teresa M Brophy
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.)
| | - James S O'Donnell
- From the Haemostasis Research Group, Institute of Molecular Medicine, Trinity Centre for Health Sciences (J.M.O., P.V.J., O.R., K.G., A.C., M.L., T.M.B., J.S.O.) and National Centre for Hereditary Coagulation Disorders (J.S.O.), St. James's Hospital, and Department of Clinical Medicine, School of Medicine (R.J.S.P.), Trinity College, Dublin, Ireland; School of Biotechnology and Biomedical Diagnostics Institute, Dublin City University, Dublin, Ireland (B.B., R.O.); and National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland (R.J.S.P.).
| |
Collapse
|
57
|
Jourdy Y, Nougier C, Roualdes O, Fretigny M, Durand B, Negrier C, Vinciguerra C. Characterization of five associations ofF8missense mutations containing FVIII B domain mutations. Haemophilia 2016; 22:583-9. [DOI: 10.1111/hae.12906] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2015] [Indexed: 01/17/2023]
Affiliation(s)
- Y. Jourdy
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
- EAM 4174 Hémostase; Inflammation et Sepsis; Université Claude Bernard Lyon1; Université de Lyon; Lyon France
| | - C. Nougier
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
- EAM 4174 Hémostase; Inflammation et Sepsis; Université Claude Bernard Lyon1; Université de Lyon; Lyon France
| | - O. Roualdes
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
- EAM 4174 Hémostase; Inflammation et Sepsis; Université Claude Bernard Lyon1; Université de Lyon; Lyon France
| | - M. Fretigny
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
| | - B. Durand
- Hospices Civils de Lyon; Hôpital de la Croix Rousse; Service d'hématologie Biologique; Lyon France
| | - C. Negrier
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
- EAM 4174 Hémostase; Inflammation et Sepsis; Université Claude Bernard Lyon1; Université de Lyon; Lyon France
| | - C. Vinciguerra
- Hospices Civils de Lyon; Hôpital Edouard Herriot; Service d'hématologie Biologique; Lyon France
- EAM 4174 Hémostase; Inflammation et Sepsis; Université Claude Bernard Lyon1; Université de Lyon; Lyon France
| |
Collapse
|
58
|
Mathew P, Dinter H, Church N, Humphries TJ, Kulkarni R. Inhibitors in haemophilia A: a perspective on clotting factor products as a potential contributing factor. Haemophilia 2016; 22:334-41. [PMID: 26843214 DOI: 10.1111/hae.12888] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2015] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The occurrence of a neutralizing antibody in previously untreated patients (PUPs) with haemophilia A appears to be the result of an intricate interplay of both genetic and environmental factors. Recently, the type of factor VIII (FVIII) product used in the PUPs population has been implicated as a risk factor for inhibitor development. AIM The aim of this review was to explore in a systematic manner potential hypotheses for the product-related findings in these studies (i.e. differences in the expression system of the cell lines used to produce recombinant FVIII [rFVIII], differences in the administered antigen load or changes in clinical practice over time). RESULTS Review of the available clinical studies illustrates the high degree of variability for the risk of inhibitor development for the same products across different studies. Differences in cell lines or antigen load were not found to provide a reasonable explanation. CONCLUSION The possibility of changes in clinical practice over time and patient selection bias (i.e. the preferential use of one product over another in patients at higher risk for inhibitors) offers a potential explanation and should be carefully considered when evaluating the studies.
Collapse
Affiliation(s)
- P Mathew
- Bayer HealthCare Pharmaceuticals Inc., Whippany, NJ, USA
| | - H Dinter
- Bayer Pharma AG, Leverkusen, Germany
| | - N Church
- Bayer HealthCare Pharmaceuticals Inc., Whippany, NJ, USA
| | - T J Humphries
- Bayer HealthCare Pharmaceuticals Inc., Whippany, NJ, USA
| | - R Kulkarni
- Michigan State University, East Lansing, MI, USA
| |
Collapse
|
59
|
Csencsits-Smith K, Grushin K, Stoilova-McPhie S. Binding of Factor VIII to Lipid Nanodiscs Increases its Clotting Function in a Mouse Model of Hemophilia A. ACTA ACUST UNITED AC 2015; 6:325. [PMID: 28936365 PMCID: PMC5603210 DOI: 10.4172/2155-9864.1000325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Hemophilia A is a congenital bleeding disorder caused by defective or deficient factor VIII (FVIII). The active form of FVIII is the co-factor for the serine protease factor IXa (FIXa) in the membrane-bound intrinsic tenase (FVIIIa-FIXa) complex. The assembly of the FVIIIa-FIXa complex on the activated platelet surface is critical for successful blood clotting. Objectives To characterize the role of lipid nanodiscs (ND) for on FVIII function in vivo and test the lipid ND as a delivery system for FVIII. To evaluate the potential of binding recombinant FVIII to ND as improved treatment for Hemophilia A. Methods Recombinant porcine FVIII (rpFVIII) was expressed and characterized in solution, and when bound to ND. The rpFVIII, ND and rpFVIII-ND complexes were characterized via transmission electron microscopy. Functional studies were carried out using aPTT tests and time resolved tail snip studies of hemophilic mice. Results Functional rpFVIII was successfully assembled on lipid ND. When injected in hemophilic mice, the rpFVIII-ND complexes showed a pronounced pro-coagulant effect, which was stronger than that of rpFVIII alone. While injection of the ND alone showed a pro-coagulant effect this effect was not additive, implying that the rpFVIII-ND complexes have a synergistic effect on the clotting process in hemophilic mice. Conclusions Binding of rpFVIII to ND prior to its injection in hemophilic mice significantly improves the therapeutic function of the protein. This represents a meaningful step towards a new approach to modulate blood coagulation at the membrane-bound FVIII level and the assembly of the intrinsic tenase complex.
Collapse
Affiliation(s)
- Keri Csencsits-Smith
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, Houston, TX-77030, USA
| | - Krill Grushin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX-77555, USA
| | - Svetla Stoilova-McPhie
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX-77555, USA
| |
Collapse
|
60
|
Wuerth ME, Cragerud RK, Clint Spiegel P. Structure of the Human Factor VIII C2 Domain in Complex with the 3E6 Inhibitory Antibody. Sci Rep 2015; 5:17216. [PMID: 26598467 PMCID: PMC4657047 DOI: 10.1038/srep17216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/27/2015] [Indexed: 01/07/2023] Open
Abstract
Blood coagulation factor VIII is a glycoprotein cofactor that is essential for the intrinsic pathway of the blood coagulation cascade. Inhibitory antibodies arise either spontaneously or in response to therapeutic infusion of functional factor VIII into hemophilia A patients, many of which are specific to the factor VIII C2 domain. The immune response is largely parsed into "classical" and "non-classical" inhibitory antibodies, which bind to opposing faces cooperatively. In this study, the 2.61 Å resolution structure of the C2 domain in complex with the antigen-binding fragment of the 3E6 classical inhibitory antibody is reported. The binding interface is largely conserved when aligned with the previously determined structure of the C2 domain in complex with two antibodies simultaneously. Further inspection of the B factors for the C2 domain in various X-ray crystal structures indicates that 3E6 antibody binding decreases the thermal motion behavior of surface loops in the C2 domain on the opposing face, thereby suggesting that cooperative antibody binding is a dynamic effect. Understanding the structural nature of the immune response to factor VIII following hemophilia A treatment will help lead to the development of better therapeutic reagents.
Collapse
Affiliation(s)
- Michelle E. Wuerth
- Western Washington University, Department of Chemistry, 516 High Street, Bellingham, WA 98225-9150
| | - Rebecca K. Cragerud
- Western Washington University, Department of Chemistry, 516 High Street, Bellingham, WA 98225-9150
| | - P. Clint Spiegel
- Western Washington University, Department of Chemistry, 516 High Street, Bellingham, WA 98225-9150,
| |
Collapse
|
61
|
Li JN, Carrero IG, Dong JF, Yu FL. Complexity and diversity of F8 genetic variations in the 1000 genomes. J Thromb Haemost 2015; 13:2031-40. [PMID: 26383047 PMCID: PMC4928474 DOI: 10.1111/jth.13144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hemophilia A (HA) is an X-linked bleeding disorder caused by deleterious mutations in the coagulation factor VIII gene (F8). To date, F8 mutations have been documented predominantly in European subjects and in American subjects of European descent. Information on F8 variants in individuals of more diverse ethnic backgrounds is limited. OBJECTIVES To discover novel and rare F8 variants, and to characterize F8 variants in diverse population backgrounds. PATIENTS/METHODS We analyzed 2535 subjects, including 26 different ethnicities, whose data were available from the 1000 Genomes Project (1000G) phase 3 dataset, for F8 variants and their potential functional impact. RESULTS We identified 3030 single nucleotide variants, 31 short deletions and insertions (Indels) and a large, 497 kb, deletion. Among all variants, 86.4% were rare variants and 55.6% were novel. Eighteen variants previously associated with HA were found in our study. Most of these 'HA variants' were ethnic-specific with low allele frequency; however, one variant (p.M2257V) was present in 27% of African subjects. The p.E132D, p.T281A, p.A303V and p.D422H 'HA variants' were identified only in males. Twelve novel missense variants were predicted to be deleterious. The large deletion was discovered in eight female subjects without affecting F8 transcription and the transcription of genes on the X chromosome. CONCLUSION Characterizing F8 in the 1000G project highlighted the complexity of F8 variants and the importance of interrogating genetic variants on multiple ethnic backgrounds for associations with bleeding and thrombosis. The haplotype analysis and the orientation of duplicons that flank the large deletion suggested that the deletion was recurrent and originated by homologous recombination.
Collapse
Affiliation(s)
- J N Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - I G Carrero
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - J F Dong
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA
- Puget Sound Blood Center, Seattle, WA, USA
| | - F L Yu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Tianjin Neurology Institute, Tianjin Medical University, Tianjin, China
| |
Collapse
|
62
|
Factor VIII Is Synthesized in Human Endothelial Cells, Packaged in Weibel-Palade Bodies and Secreted Bound to ULVWF Strings. PLoS One 2015; 10:e0140740. [PMID: 26473492 PMCID: PMC4608722 DOI: 10.1371/journal.pone.0140740] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/30/2015] [Indexed: 11/19/2022] Open
Abstract
The cellular synthesis site and ensuing storage location for human factor VIII (FVIII), the coagulation protein deficient in hemophilia A, has been elusive. FVIII stability and half-life is dependent on non-covalent complex formation with von Willebrand factor (VWF) to avoid proteolysis and clearance. VWF is synthesized in megakaryocytes and endothelial cells, and is stored and secreted from platelet alpha granules and Weibel-Palade bodies of endothelial cells. In this paper we provide direct evidence for FVIII synthesis in 2 types of primary human endothelial cells: glomerular microvascular endothelial cells (GMVECs) and umbilical vein endothelial cells (HUVECs). Gene expression quantified by real time PCR revealed that levels of F8 and VWF are similar in GMVECs and HUVECs. Previous clinical studies have shown that stimulation of vasopressin V2 receptors causes parallel secretion of both proteins. In this study, we found that both endothelial cell types express AVPR2 (vasopressin V2 receptor gene) and that AVPR2 mRNA levels are 5-fold higher in GMVECs than HUVECs. FVIII and VWF proteins were detected by fluorescent microscopy in Weibel-Palade bodies within GMVECs and HUVECs using antibodies proven to be target specific. Visual presence of FVIII and VWF in Weibel-Palade bodies was confirmed by correlation measurements. The high extent of correlation was compared with negative correlation values obtained from FVIII detection with cytoplasmic proteins, β-actin and Factor H. FVIII activity was positive in GMVEC and HUVEC cell lysates. Stimulated GMVECs and HUVECs were found to secrete cell-anchored ultra-large VWF strings covered with bound FVIII.
Collapse
|
63
|
Platelet binding sites for factor VIII in relation to fibrin and phosphatidylserine. Blood 2015; 126:1237-44. [PMID: 26162408 DOI: 10.1182/blood-2015-01-620245] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 07/07/2015] [Indexed: 11/20/2022] Open
Abstract
Thrombin-stimulated platelets expose very little phosphatidylserine (PS) but express binding sites for factor VIII (fVIII), casting doubt on the role of exposed PS as the determinant of binding sites. We previously reported that fVIII binding sites are increased three- to sixfold when soluble fibrin (SF) binds the αIIbβ3 integrin. This study focuses on the hypothesis that platelet-bound SF is the major source of fVIII binding sites. Less than 10% of fVIII was displaced from thrombin-stimulated platelets by lactadherin, a PS-binding protein, and an fVIII mutant defective in PS-dependent binding retained platelet affinity. Therefore, PS is not the determinant of most binding sites. FVIII bound immobilized SF and paralleled platelet binding in affinity, dependence on separation from von Willebrand factor, and mediation by the C2 domain. SF also enhanced activity of fVIII in the factor Xase complex by two- to fourfold. Monoclonal antibody (mAb) ESH8, against the fVIII C2 domain, inhibited binding of fVIII to SF and platelets but not to PS-containing vesicles. Similarly, mAb ESH4 against the C2 domain, inhibited >90% of platelet-dependent fVIII activity vs 35% of vesicle-supported activity. These results imply that platelet-bound SF is a component of functional fVIII binding sites.
Collapse
|
64
|
Chahal G, Thorpe M, Hellman L. The Importance of Exosite Interactions for Substrate Cleavage by Human Thrombin. PLoS One 2015; 10:e0129511. [PMID: 26110612 PMCID: PMC4482499 DOI: 10.1371/journal.pone.0129511] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/08/2015] [Indexed: 11/18/2022] Open
Abstract
Thrombin is a serine protease of the chymotrypsin family that acts both as a procoagulant and as an anticoagulant by cleaving either factor VIII, factor V and fibrinogen or protein C, respectively. Numerous previous studies have shown that electropositive regions at a distance from the active site, so called exosites, are of major importance for the cleavage by human thrombin. Upstream of all the known major cleavage sites for thrombin in factor VIII, factor V and fibrinogen are clusters of negatively charged amino acids. To study the importance of these sites for the interaction with the exosites and thereby the cleavage by thrombin, we have developed a new type of recombinant substrate. We have compared the cleavage rate of the minimal cleavage site, involving only 8-9 amino acids (typically the P4-P4' positions) surrounding the cleavage site, with the substrates also containing the negatively charged regions upstream of the cleavage sites. The results showed that addition of these regions enhanced the cleavage rate by more than fifty fold. However, the enhancement was highly dependent on the sequence of the actual cleavage site. A minimal site that showed poor activity by itself could be cleaved as efficiently as an optimal cleavage site when presented together with these negatively charged regions. Whereas sites conforming closely to the optimal site were only minimally enhanced by the addition of these regions. The possibility to mimic this interaction for the sites in factor V and factor VIII by recombinant substrates, which do not have the same folding as the full size target, indicates that the enhancement was primarily dependent on a relatively simple electrostatic interaction. However, the situation was very different for fibrinogen and protein C where other factors than only charge is of major importance.
Collapse
Affiliation(s)
- Gurdeep Chahal
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden
| | - Michael Thorpe
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, BMC, Box 596, SE-751 24, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
65
|
Dalm D, Galaz-Montoya JG, Miller JL, Grushin K, Villalobos A, Koyfman AY, Schmid MF, Stoilova-McPhie S. Dimeric Organization of Blood Coagulation Factor VIII bound to Lipid Nanotubes. Sci Rep 2015; 5:11212. [PMID: 26082135 PMCID: PMC4469981 DOI: 10.1038/srep11212] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/05/2015] [Indexed: 11/09/2022] Open
Abstract
Membrane-bound Factor VIII (FVIII) has a critical function in blood coagulation as the pro-cofactor to the serine-protease Factor IXa (FIXa) in the FVIIIa-FIXa complex assembled on the activated platelet membrane. Defects or deficiency of FVIII cause Hemophilia A, a mild to severe bleeding disorder. Despite existing crystal structures for FVIII, its membrane-bound organization has not been resolved. Here we present the dimeric FVIII membrane-bound structure when bound to lipid nanotubes, as determined by cryo-electron microscopy. By combining the structural information obtained from helical reconstruction and single particle subtomogram averaging at intermediate resolution (15-20 Å), we show unambiguously that FVIII forms dimers on lipid nanotubes. We also demonstrate that the organization of the FVIII membrane-bound domains is consistently different from the crystal structure in solution. The presented results are a critical step towards understanding the mechanism of the FVIIIa-FIXa complex assembly on the activated platelet surface in the propagation phase of blood coagulation.
Collapse
Affiliation(s)
- Daniela Dalm
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jesus G Galaz-Montoya
- 1] Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA [2] National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jaimy L Miller
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kirill Grushin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alex Villalobos
- School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alexey Y Koyfman
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Michael F Schmid
- 1] Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA [2] National Center for Macromolecular Imaging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Svetla Stoilova-McPhie
- 1] Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA [2] Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
66
|
The 1.7 Å X-ray crystal structure of the porcine factor VIII C2 domain and binding analysis to anti-human C2 domain antibodies and phospholipid surfaces. PLoS One 2015; 10:e0122447. [PMID: 25775247 PMCID: PMC4361576 DOI: 10.1371/journal.pone.0122447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
The factor VIII C2 domain is essential for binding to activated platelet surfaces as well as the cofactor activity of factor VIII in blood coagulation. Inhibitory antibodies against the C2 domain commonly develop following factor VIII replacement therapy for hemophilia A patients, or they may spontaneously arise in cases of acquired hemophilia. Porcine factor VIII is an effective therapeutic for hemophilia patients with inhibitor due to its low cross-reactivity; however, the molecular basis for this behavior is poorly understood. In this study, the X-ray crystal structure of the porcine factor VIII C2 domain was determined, and superposition of the human and porcine C2 domains demonstrates that most surface-exposed differences cluster on the face harboring the "non-classical" antibody epitopes. Furthermore, antibody-binding results illustrate that the "classical" 3E6 antibody can bind both the human and porcine C2 domains, although the inhibitory titer to human factor VIII is 41 Bethesda Units (BU)/mg IgG versus 0.8 BU/mg IgG to porcine factor VIII, while the non-classical G99 antibody does not bind to the porcine C2 domain nor inhibit porcine factor VIII activity. Further structural analysis of differences between the electrostatic surface potentials suggest that the C2 domain binds to the negatively charged phospholipid surfaces of activated platelets primarily through the 3E6 epitope region. In contrast, the G99 face, which contains residue 2227, should be distal to the membrane surface. Phospholipid binding assays indicate that both porcine and human factor VIII C2 domains bind with comparable affinities, and the human K2227A and K2227E mutants bind to phospholipid surfaces with similar affinities as well. Lastly, the G99 IgG bound to PS-immobilized factor VIII C2 domain with an apparent dissociation constant of 15.5 nM, whereas 3E6 antibody binding to PS-bound C2 domain was not observed.
Collapse
|
67
|
Pan TY, Chiou SS, Wang CC, Wu SM. Separation of intron 22 inversion type 1 and 2 of hemophilia A by modified inverse-shifting polymerase chain reaction and capillary gel electrophoresis. Talanta 2014; 130:328-35. [DOI: 10.1016/j.talanta.2014.06.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/28/2014] [Accepted: 06/30/2014] [Indexed: 10/25/2022]
|
68
|
Fomin ME, Togarrati PP, Muench MO. Progress and challenges in the development of a cell-based therapy for hemophilia A. J Thromb Haemost 2014; 12:1954-65. [PMID: 25297648 PMCID: PMC4388483 DOI: 10.1111/jth.12750] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Indexed: 12/11/2022]
Abstract
Hemophilia A results from an insufficiency of factor VIII (FVIII). Although replacement therapy with plasma-derived or recombinant FVIII is a life-saving therapy for hemophilia A patients, such therapy is a life-long treatment rather than a cure for the disease. In this review, we discuss the possibilities, progress, and challenges that remain in the development of a cell-based cure for hemophilia A. The success of cell therapy depends on the type and availability of donor cells, the age of the host and method of transplantation, and the levels of engraftment and production of FVIII by the graft. Early therapy, possibly even prenatal transplantation, may yield the highest levels of engraftment by avoiding immunological rejection of the graft. Potential cell sources of FVIII include a specialized subset of endothelial cells known as liver sinusoidal endothelial cells (LSECs) present in the adult and fetal liver, or patient-specific endothelial cells derived from induced pluripotent stem cells that have undergone gene editing to produce FVIII. Achieving sufficient engraftment of transplanted LSECs is one of the obstacles to successful cell therapy for hemophilia A. We discuss recent results from transplants performed in animals that show production of functional and clinically relevant levels of FVIII obtained from donor LSECs. Hence, the possibility of treating hemophilia A can be envisioned through persistent production of FVIII from transplanted donor cells derived from a number of potential cell sources or through creation of donor endothelial cells from patient-specific induced pluripotent stem cells.
Collapse
Affiliation(s)
- Marina E. Fomin
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Padma Priya Togarrati
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
| | - Marcus O. Muench
- Cell Therapy Core, Blood Systems Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California, San Francisco, CA
- Liver Center, University of California, San Francisco, CA
| |
Collapse
|
69
|
Abstract
BACKGROUND Haemophilia is a genetic disorder which is characterized by spontaneous or provoked, often uncontrolled, bleeding into joints, muscles and other soft tissues. Current methods of treatment are expensive, challenging and involve regular administration of clotting factors. Gene therapy has recently been prompted as a curative treatment modality. OBJECTIVES To evaluate the safety and efficacy of gene therapy for treating people with haemophilia A or B. SEARCH METHODS We searched the Cochrane Cystic Fibrosis & Genetic Disorders Group's Coagulopathies Trials Register, compiled from electronic database searches and handsearching of journals and conference abstract books. We also searched the reference lists of relevant articles and reviews.Date of last search: 06 November 2014. SELECTION CRITERIA Eligible trials included randomised or quasi-randomised clinical trials, including controlled clinical trials comparing gene therapy (with or without standard treatment) with standard treatment (factor replacement) or other 'curative' treatment such as stem cell transplantation individuals with haemophilia A or B of all ages who do not have inhibitors to factor VIII or IX. DATA COLLECTION AND ANALYSIS No trials of gene therapy for haemophilia were found. MAIN RESULTS No trials of gene therapy for haemophilia were identified. AUTHORS' CONCLUSIONS No randomised or quasi-randomised clinical trials of gene therapy for haemophilia were identified. Thus, we are unable to determine the effects of gene therapy for haemophilia. Gene therapy for haemophilia is still in its nascent stages and there is a need for well-designed clinical trials to assess the long-term feasibility, success and risks of gene therapy for people with haemophilia.
Collapse
Affiliation(s)
- Akshay Sharma
- Department of Pediatrics, University of Kentucky, 800 Rose Street, Room MN118, Lexington, Kentucky, USA, 40536
| | | | | | | | | |
Collapse
|
70
|
Mohammadian T, Rassi H. Investigation of hFVIII production in mammary glands of transgenic mice. Monoclon Antib Immunodiagn Immunother 2014; 33:325-9. [PMID: 25358000 DOI: 10.1089/mab.2014.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hemophilia A is an X-linked disorder affecting 1 in 10,000 males. The disease is caused by a defect or mutation of factor 8 or 9. Human factor 8 gene (hFVIII) is a relatively large gene consisting of 26 exons and approximately 2,351 amino acids with a length of 9 Kb mRNA. Expression of hFVIII in mammalian milk is becoming a widespread strategy for high-level production of hFVIII because of the most complex post-translational modifications. The aim of this study was the cloning and expression of hFVIII in mammary glands of two transgenic mice. To obtain a recombinant plasmid, first a plasmid carrying an FVIII gene fragment (pCMV6-hFVIII) was digested by EcoRI-SalI restriction enzymes and then the fragment was purified from agarose gel and inserted into a pUCWAP7 vector carrying a tissue-specific promoter (mWAP 4.1 kbp). After that, it was isolated by agarose gel and transferred into the murine zygotes by standard microinjection methods. Methods for expression of recombinant FVIII RT-PCR and ELISA were studied. The results show the successful expression of factor FVIII gene and its product in the mouse mammary glands.
Collapse
Affiliation(s)
- Tahar Mohammadian
- 1 Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University , Tehran, Iran
| | | |
Collapse
|
71
|
Changes in the Factor VIII C2 domain upon membrane binding determined by hydrogen–deuterium exchange MS. Biochem J 2014; 461:443-51. [DOI: 10.1042/bj20140121] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Blood coagulation Factor VIII binds to a membrane in order to function as a cofactor for Factor IXa, preventing haemophilia. The present study indicates that membrane-binding peptides of Factor VIII are largely protected from water exposure, indicating that they become immersed in the membrane.
Collapse
|
72
|
Miller J, Dalm D, Koyfman AY, Grushin K, Stoilova-McPhie S. Helical organization of blood coagulation factor VIII on lipid nanotubes. J Vis Exp 2014. [PMID: 24961276 DOI: 10.3791/51254] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cryo-electron microscopy (Cryo-EM)(1) is a powerful approach to investigate the functional structure of proteins and complexes in a hydrated state and membrane environment(2). Coagulation Factor VIII (FVIII)(3) is a multi-domain blood plasma glycoprotein. Defect or deficiency of FVIII is the cause for Hemophilia type A - a severe bleeding disorder. Upon proteolytic activation, FVIII binds to the serine protease Factor IXa on the negatively charged platelet membrane, which is critical for normal blood clotting(4). Despite the pivotal role FVIII plays in coagulation, structural information for its membrane-bound state is incomplete(5). Recombinant FVIII concentrate is the most effective drug against Hemophilia type A and commercially available FVIII can be expressed as human or porcine, both forming functional complexes with human Factor IXa(6,7). In this study we present a combination of Cryo-electron microscopy (Cryo-EM), lipid nanotechnology and structure analysis applied to resolve the membrane-bound structure of two highly homologous FVIII forms: human and porcine. The methodology developed in our laboratory to helically organize the two functional recombinant FVIII forms on negatively charged lipid nanotubes (LNT) is described. The representative results demonstrate that our approach is sufficiently sensitive to define the differences in the helical organization between the two highly homologous in sequence (86% sequence identity) proteins. Detailed protocols for the helical organization, Cryo-EM and electron tomography (ET) data acquisition are given. The two-dimensional (2D) and three-dimensional (3D) structure analysis applied to obtain the 3D reconstructions of human and porcine FVIII-LNT is discussed. The presented human and porcine FVIII-LNT structures show the potential of the proposed methodology to calculate the functional, membrane-bound organization of blood coagulation Factor VIII at high resolution.
Collapse
Affiliation(s)
- Jaimy Miller
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch
| | - Daniela Dalm
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch
| | - Alexey Y Koyfman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch
| | - Kirill Grushin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch
| | - Svetla Stoilova-McPhie
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch;
| |
Collapse
|
73
|
Ezban M, Vad K, Kjalke M. Turoctocog alfa (NovoEight®)--from design to clinical proof of concept. Eur J Haematol 2014; 93:369-76. [PMID: 24797664 PMCID: PMC4232928 DOI: 10.1111/ejh.12366] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2014] [Indexed: 11/29/2022]
Abstract
Turoctocog alfa (NovoEight®) is a recombinant factor VIII (rFVIII) with a truncated B-domain made from the sequence coding for 10 amino acids from the N-terminus and 11 amino acids from the C-terminus of the naturally occurring B-domain. Turoctocog alfa is produced in Chinese hamster ovary (CHO) cells without addition of any human- or animal-derived materials. During secretion, some rFVIII molecules are cleaved at the C-terminal of the heavy chain (HC) at amino acid 720, and a monoclonal antibody binding C-terminal to this position is used in the purification process allowing isolation of the intact rFVIII. Viral inactivation is ensured by a detergent inactivation step as well as a 20-nm nano-filtration step. Characterisation of the purified protein demonstrated that turoctocog alfa was fully sulphated at Tyr346 and Tyr1664, which is required for optimal proteolytic activation by thrombin. Kinetic assessments confirmed that turoctocog alfa was activated by thrombin at a similar rate as seen for other rFVIII products fully sulphated at these positions. Tyr1680 was also fully sulphated in turoctocog alfa resulting in strong affinity (low nm Kd) for binding to von Willebrand factor (VWF). Half-lives of 7.2 ± 0.9 h in F8-KO mice and 8.9 ± 1.8 h haemophilia A dogs supported that turoctocog alfa bound to VWF after infusion. Functional studies including thromboelastography analysis of human haemophilia A whole blood with added turoctocog alfa and effect studies in mice bleeding models demonstrated a dose-dependent effect of turoctocog alfa. The non-clinical data thus confirm the haemostatic effect of turoctocog alfa and, together with the comprehensive clinical evaluation, support the use as FVIII replacement therapy in patients with haemophilia A.
Collapse
|
74
|
Grushin K, Miller J, Dalm D, Parker ET, Healey JF, Lollar P, Stoilova-McPhie S. Lack of recombinant factor VIII B-domain induces phospholipid vesicle aggregation: implications for the immunogenicity of factor VIII. Haemophilia 2014; 20:723-31. [PMID: 24750465 PMCID: PMC4149818 DOI: 10.1111/hae.12421] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2014] [Indexed: 11/28/2022]
Abstract
Factor VIII (FVIII) is a multidomain blood plasma glycoprotein. Activated FVIII acts as a cofactor to the serine protease factor IXa within the membrane-bound tenase complex assembled on the activated platelet surface. Defect or deficiency in FVIII causes haemophilia A, a severe hereditary bleeding disorder. Intravenous administration of plasma-derived FVIII or recombinant FVIII concentrates restores normal coagulation in haemophilia A patients and is used as an effective therapy. In this work, we studied the biophysical properties of clinically potent recombinant FVIII forms: human FVIII full-length (FVIII-FL), human FVIII B-domain deleted (FVIII-BDD) and porcine FVIII-BDD bound to negatively charged phospholipid vesicles at near-physiological conditions. We used cryo-electron microscopy (Cryo-EM) as a direct method to evaluate the homogeneity and micro-organization of the protein-vesicle suspensions, which are important for FVIII therapeutic properties. Applying concurrent Cryo-EM, circular dichroism and dynamic light scattering studies to the three recombinant FVIII forms when bound to phospholipid vesicles revealed novel properties for their functional, membrane-bound state. The three FVIII constructs have similar activity, secondary structure distribution and bind specifically to negatively charged phospholipid membranes. Human and porcine FVIII-BDD induce strong aggregation of the vesicles, but the human FVIII-FL form does not. The proposed methodology is effective in characterizing and identifying differences in therapeutic recombinant FVIII membrane-bound forms near physiological conditions, because protein-containing aggregates are considered to be a factor in increasing the immunogenicity of protein therapeutics. This will provide better characterization and development of safer and more effective FVIII products with implications for haemophilia A treatment.
Collapse
Affiliation(s)
- K Grushin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Aledort L, Ljung R, Mann K, Pipe S. Factor VIII therapy for hemophilia A: current and future issues. Expert Rev Hematol 2014; 7:373-85. [PMID: 24717090 DOI: 10.1586/17474086.2014.899896] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Hemophilia A is a congenital, recessive, X-linked bleeding disorder that is managed with infusions of plasma-derived or recombinant factor (F) VIII. The primary considerations in FVIII replacement therapy today are the: 1) immunogenicity of FVIII concentrates, 2) role of longer-acting FVIII products, 3) prophylactic use of FVIII in children and adults with severe hemophilia A, and 4) affordability and availability of FVIII products. Improving patient outcomes by increasing the use of FVIII prophylaxis, preventing or eliminating FVIII inhibitors, and expanding access to FVIII concentrates in developing countries are the major challenges confronting clinicians who care for patients with hemophilia A.
Collapse
Affiliation(s)
- Louis Aledort
- Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1006, Newyork, NY, USA
| | | | | | | |
Collapse
|
76
|
Mikhail S, Aldin ES, Streiff M, Zeidan A. An update on type 2B von Willebrand disease. Expert Rev Hematol 2014; 7:217-31. [PMID: 24521271 DOI: 10.1586/17474086.2014.868771] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Type 2B von Willebrand disease (VWD) accounts for fewer than 5% of all VWD patients. In this disease, mutations in the A1 domain result in increased von Willebrand factor (VWF) binding to platelet GPIbα receptors, causing increased platelet clearance and preferential loss of high molecular weight VWF multimers. Diagnosis is complicated because of significant clinical variations even among patients with identical mutations. Platelet transfusion often provides suboptimal results since transfused platelets may be aggregated by the patients' abnormal VWF. Desmopressin may cause a transient decrease in platelet count that could lead to an increased risk of bleeding. Replacement therapy with factor VIII/VWF concentrates is the most effective approach to prevention and treatment of bleeding in type 2B VWD.
Collapse
Affiliation(s)
- Sameh Mikhail
- Department of Hematology, Ohio State University Medical Center, Columbus, OH, USA
| | | | | | | |
Collapse
|
77
|
Cugno M, Gualtierotti R, Tedeschi A, Meroni PL. Autoantibodies to coagulation factors: From pathophysiology to diagnosis and therapy. Autoimmun Rev 2014; 13:40-8. [DOI: 10.1016/j.autrev.2013.08.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 12/20/2022]
|
78
|
Griffiths AE, Wintermute J, Newell-Caito JL, Fay PJ. Residues flanking scissile bonds in Factor VIII modulate rates of cleavage and proteolytic activation catalyzed by Factor Xa. Biochemistry 2013; 52:8060-8. [PMID: 24128092 DOI: 10.1021/bi4010123] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Factor Xa (FXa) proteolytically activates Factor VIII (FVIII) by cleaving P1 residues Arg(372), Arg(740), and Arg(1689). The Arg(372) site represents the rate-limiting step for procofactor activation, whereas cleavage at Arg(740) is a fast step. FXa also catalyzes inactivating cleavages that occur on a slower time scale than the activating ones. To assess the role of sequences flanking the Arg(372) and Arg(740) sites, recombinant FVIII variants in which P3-P3' sequences were swapped individually or in combination were prepared. Replacing the Arg(372) flanking sequence with that from the Arg(740) site increased the rate of cleavage at Arg(372), as judged by the ~5-fold increased rate in A1 subunit generation, and reduced the FVIIIa-dependent lag time for in situ FXa generation. The reciprocal swap yielded a nearly 2-fold increase in the rate of Arg(372) cleavage, while the combined double-swap variant showed a 10-fold rate increase at that site, consistent with the individual effects being additive. Although this cleavage represents the slow step for activation, the rate of this reaction appeared to be ~9-fold greater than the rate of the primary inactivating cleavage at Arg(336) in generating the A1(336) product. Interestingly, replacement of the Arg(372) flanking sequence with the Arg(740) sequence combined with an Arg(740)Gln mutation yielded both more rapid cleavage of the Arg(372) site and accelerated inactivating cleavages within the A1 subunit. These results indicate that flanking sequences in part modulate the reaction rates required for procofactor activation and influence the capacity of FXa as an initial activator of FVIII rather than an inactivator.
Collapse
Affiliation(s)
- Amy E Griffiths
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine , 601 Elmwood Avenue, Rochester, New York 14642, United States
| | | | | | | |
Collapse
|
79
|
Structure of the factor VIII C2 domain in a ternary complex with 2 inhibitor antibodies reveals classical and nonclassical epitopes. Blood 2013; 122:4270-8. [PMID: 24085769 DOI: 10.1182/blood-2013-08-519124] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The factor VIII C2 domain is a highly immunogenic domain, whereby inhibitory antibodies develop following factor VIII replacement therapy for congenital hemophilia A patients. Inhibitory antibodies also arise spontaneously in cases of acquired hemophilia A. The structural basis for molecular recognition by 2 classes of anti-C2 inhibitory antibodies that bind to factor VIII simultaneously was investigated by x-ray crystallography. The C2 domain/3E6 FAB/G99 FAB ternary complex illustrates that each antibody recognizes epitopes on opposing faces of the factor VIII C2 domain. The 3E6 epitope forms direct contacts to the C2 domain at 2 loops consisting of Glu2181-Ala2188 and Thr2202-Arg2215, whereas the G99 epitope centers on Lys2227 and also makes direct contacts with loops Gln2222-Trp2229, Leu2261-Ser2263, His2269-Val2282, and Arg2307-Gln2311. Each binding interface is highly electrostatic, with positive charge present on both C2 epitopes and complementary negative charge on each antibody. A new model of membrane association is also presented, where the 3E6 epitope faces the negatively charged membrane surface and Arg2320 is poised at the center of the binding interface. These results illustrate the potential complexities of the polyclonal anti-factor VIII immune response and further define the "classical" and "nonclassical" types of antibody inhibitors against the factor VIII C2 domain.
Collapse
|
80
|
Stoilova-McPhie S, Lynch GC, Ludtke S, Pettitt BM. Domain organization of membrane-bound factor VIII. Biopolymers 2013; 99:448-59. [PMID: 23616213 PMCID: PMC4090243 DOI: 10.1002/bip.22199] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/13/2012] [Accepted: 11/18/2012] [Indexed: 11/07/2022]
Abstract
Factor VIII (FVIII) is the blood coagulation protein which when defective or deficient causes for hemophilia A, a severe hereditary bleeding disorder. Activated FVIII (FVIIIa) is the cofactor to the serine protease factor IXa (FIXa) within the membrane-bound Tenase complex, responsible for amplifying its proteolytic activity more than 100,000 times, necessary for normal clot formation. FVIII is composed of two noncovalently linked peptide chains: a light chain (LC) holding the membrane interaction sites and a heavy chain (HC) holding the main FIXa interaction sites. The interplay between the light and heavy chains (HCs) in the membrane-bound state is critical for the biological efficiency of FVIII. Here, we present our cryo-electron microscopy (EM) and structure analysis studies of human FVIII-LC, when helically assembled onto negatively charged single lipid bilayer nanotubes. The resolved FVIII-LC membrane-bound structure supports aspects of our previously proposed FVIII structure from membrane-bound two-dimensional (2D) crystals, such as only the C2 domain interacts directly with the membrane. The LC is oriented differently in the FVIII membrane-bound helical and 2D crystal structures based on EM data, and the existing X-ray structures. This flexibility of the FVIII-LC domain organization in different states is discussed in the light of the FVIIIa-FIXa complex assembly and function.
Collapse
Affiliation(s)
- Svetla Stoilova-McPhie
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | | | |
Collapse
|
81
|
Peyvandi F, Garagiola I, Seregni S. Future of coagulation factor replacement therapy. J Thromb Haemost 2013; 11 Suppl 1:84-98. [PMID: 23809113 DOI: 10.1111/jth.12270] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/13/2013] [Indexed: 12/24/2022]
Abstract
Over a million patients worldwide currently suffer from hemophilia and other congenital clotting factor deficiencies. Patients affected with hemophilia A and B are treated by intravenous replacement therapy of factor VIII and factor IX, respectively. Current hemophilia treatments have favorably supported their efficacy, tolerability, and safety profiles. The onset of alloantibodies inactivating the infused coagulation factor is the main problem in hemophilia patients rendering replacement therapies ineffective; another disadvantage is the short half-life of the infused clotting factors with the need for multiple and frequent infusions to manage a bleeding episode. Now, the challenge in the management of hemophilia treatment is the prolongation of the half-life and reduction in the immunogenicity of recombinant clotting factors. The bioengineering strategies, previously applied successfully to other therapeutic proteins, encourage the current efforts to produce novel coagulation factors with more prolonged bioavailability, with increased potency and resistance to inactivation and potentially reduced immunogenicity.
Collapse
Affiliation(s)
- F Peyvandi
- Angelo Bianchi Bonomi Haemophilia and Thrombosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | | | | |
Collapse
|
82
|
Inaba H, Shinozawa K, Seita I, Otaki M, Suzuki T, Hagiwara T, Amano K, Fukutake K. Genotypic and phenotypic features of Japanese patients with mild to moderate hemophilia A. Int J Hematol 2013; 97:758-64. [DOI: 10.1007/s12185-013-1341-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/12/2013] [Accepted: 04/15/2013] [Indexed: 11/28/2022]
|
83
|
Walter JD, Werther RA, Polozova MS, Pohlman J, Healey JF, Meeks SL, Lollar P, Spiegel PC. Characterization and solution structure of the factor VIII C2 domain in a ternary complex with classical and non-classical inhibitor antibodies. J Biol Chem 2013; 288:9905-9914. [PMID: 23417672 DOI: 10.1074/jbc.m112.424564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The most significant complication for patients with severe cases of congenital or acquired hemophilia A is the development of inhibitor antibodies against coagulation factor VIII (fVIII). The C2 domain of fVIII is a significant antigenic target of anti-fVIII antibodies. Here, we have utilized small angle x-ray scattering (SAXS) and biochemical techniques to characterize interactions between two different classes of anti-C2 domain inhibitor antibodies and the isolated C2 domain. Multiple assays indicated that antibodies 3E6 and G99 bind independently to the fVIII C2 domain and can form a stable ternary complex. SAXS-derived numerical estimates of dimensional parameters for all studied complexes agree with the proportions of the constituent proteins. Ab initio modeling of the SAXS data results in a long kinked structure of the ternary complex, showing an angle centered at the C2 domain of ∼130°. Guided by biochemical data, rigid body modeling of subunits into the molecular envelope of the ternary complex suggests that antibody 3E6 recognizes a C2 domain epitope consisting of the Arg(2209)-Ser(2216) and Leu(2178)-Asp(2187) loops. In contrast, antibody G99 recognizes the C2 domain primarily through the Pro(2221)-Trp(2229) loop. These two epitopes are on opposing sides of the fVIII C2 domain, are consistent with the solvent accessibility in the context of the entire fVIII molecule, and provide further structural detail regarding the pathogenic immune response to fVIII.
Collapse
Affiliation(s)
- Justin D Walter
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225
| | - Rachel A Werther
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225
| | - Maria S Polozova
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225
| | - Julie Pohlman
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225
| | - John F Healey
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Shannon L Meeks
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University, Atlanta, Georgia 30322
| | - P Clint Spiegel
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225.
| |
Collapse
|
84
|
Mao J, Xi X, Kapranov P, Dong B, Firrman J, Xu R, Xiao W. In vitro and In vivo Model Systems for Hemophilia A Gene Therapy. ACTA ACUST UNITED AC 2013; Suppl 1. [PMID: 25401041 PMCID: PMC4229687 DOI: 10.4172/2157-7412.s1-014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hemophilia A is a hereditary disorder caused by various mutations in factor VIII gene resulting in either a severe deficit or total lack of the corresponding activity. Recent success in gene therapy of a related disease, hemophilia B, gives new hope that similar success can be achieved for hemophilia A as well. To develop a gene therapy strategy for the latter, a variety of model systems are needed to evaluate molecular engineering of the factor VIII gene, vector delivery efficacy and safety-related issues. Typically, a tissue culture cell line is the most convenient way to get a preliminary glimpse of the potential of a vector delivery strategy. It is then followed by extensive testing in hemophilia A mouse and dog models. Newly developed hemophilia A sheep may provide yet another tool for evaluation of factor VIII gene delivery vectors. Hemophilia models based on other species may also be developed since hemophiliac animals have been identified or generated in rat, pig, cattle and horse. Although a genetic nonhuman primate hemophilia A model has yet to be developed, the non-genetic hemophilia A model can also be used for special purposes when specific questions need to be addressed that cannot not be answered in other model systems. Hemophilia A is caused by a functional deficiency in the factor VIII gene. This X-linked, recessive bleeding disorder affects approximately 1 in 5000 males [1–3]. Clinically, it is characterized by frequent and spontaneous joint hemorrhages, easy bruising and prolonged bleeding time. The coagulation activity of FVIII dictates severity of the clinical symptoms. Approximately 50% of all cases are classified as severe with less than 1% of normal levels of factor VIII detected [4]. This deficiency may lead to spontaneous joint hemorrhages or life-threatening bleeding. In contrast, patients with 5–30% of normal factor VIII activity exhibit mild clinical manifestations.
Collapse
Affiliation(s)
- Jianhua Mao
- Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China ; Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, PA, USA
| | - Xiaodong Xi
- Shanghai Institute of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | - Biao Dong
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, PA, USA
| | - Jenni Firrman
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, PA, USA
| | - Ruian Xu
- Institute of Molecular Medicine, Molecular Medicine Engineering Research Center, Huaqiao University, Quanzhou 362021, China
| | - Weidong Xiao
- Department of Microbiology and Immunology, Sol Sherry Thrombosis Research Center, Temple University, Philadelphia, PA, USA
| |
Collapse
|
85
|
Arai I, Ohta M, Suzuki A, Tanaka S, Yoshizawa Y, Sato R. Immunohistochemical analysis of the role of hemocytin in nodule formation in the larvae of the silkworm, Bombyx mori. JOURNAL OF INSECT SCIENCE (ONLINE) 2013; 13:125. [PMID: 24766322 PMCID: PMC4015701 DOI: 10.1673/031.013.12501] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Hemocytin, a multidomain protein from Bombyx mori L. (Lepidoptera: Bombycidae), is an ortholog of von Willebrand factor and is expected to be a major mediator of hemocyte aggregation. Antiserum was generated against hemocytin, and immune staining of hemocytes, hemolymph, and nodules was performed. Hemocytin was observed in steady-state hemocytes but not in plasma, even after bacterial injection. When hemolymph was smeared on glass slides, hemocytin-containing fibrous structures formed a cellular network mainly consisting of granulocytes and oenocytoids. Hemocytin was stained only in the granules of the granulocytes. When nodule-like aggregates formed 30 sec after bacterial injection, both granulocytes and bacterial cells were observed binding to hemocytin-containing fibrous structures. When nodule sections were stained with antiserum, hemocytin was seen in the matrix of the nodules surrounding the hemocytes. These data suggest that hemocytin plays a major role in nodule formation as a component of the sticky fibrous structure exocytosed from granulocytes.
Collapse
Affiliation(s)
- Ikuyo Arai
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| | - Masayuki Ohta
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| | - Asahi Suzuki
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| | - Shiho Tanaka
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| | - Yasutaka Yoshizawa
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| | - Ryoichi Sato
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Koganei 2-24-16, Tokyo 184-8588, Japan
| |
Collapse
|
86
|
Pahl S, Pavlova A, Driesen J, Müller J, Pötzsch B, Oldenburg J. In vitrocharacterization of recombinant factor VIII concentrates reveals significant differences in protein content, activity and thrombin activation profile. Haemophilia 2012; 19:392-8. [DOI: 10.1111/hae.12076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2012] [Indexed: 11/28/2022]
Affiliation(s)
- S. Pahl
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| | - A. Pavlova
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| | - J. Driesen
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| | - J. Müller
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| | - B. Pötzsch
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| | - J. Oldenburg
- Institute of Experimental Haematology and Transfusion Medicine; University Clinic Bonn; Bonn; Germany
| |
Collapse
|
87
|
Sandberg H, Kannicht C, Stenlund P, Dadaian M, Oswaldsson U, Cordula C, Walter O. Functional characteristics of the novel, human-derived recombinant FVIII protein product, human-cl rhFVIII. Thromb Res 2012; 130:808-17. [PMID: 23010293 DOI: 10.1016/j.thromres.2012.08.311] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 08/14/2012] [Accepted: 08/28/2012] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Hemophilia A is routinely treated by administration of exogenous coagulation factor VIII (FVIII). As safety and efficacy of FVIII products have improved over the years, development of FVIII-neutralizing antibodies (FVIII inhibitors) has emerged as the most serious complication. The new human cell line-derived recombinant human FVIII (human-cl rhFVIII) is the first recombinant FVIII product produced in a human cell line without additive animal proteins, with a goal of minimizing the risk of inhibitor development. MATERIALS AND METHODS Biochemical analyzes of purity, molecular and functional attributes of the novel human-cl rhFVIII were undertaken for product characterization. RESULTS AND CONCLUSIONS Human-cl rhFVIII was shown to be highly pure, with host-cell protein and DNA traces comparable to, or lower than, currently marketed recombinant FVIII (rFVIII) products. Human-cl rhFVIII was shown to have high specific FVIII activity and characteristics similar to full-length rFVIII products. Furthermore, no significant discrepancy between one-stage and chromogenic assay results were observed for human-cl rhFVIII, indicating potency ratios of these assays comparable to the full-length rFVIII products. In functional tests, human-cl rhFVIII exhibited physiological thrombin generation and a normal rate of inactivation by activated protein C. Importantly, human-cl rhFVIII displayed higher binding capacity with von Willebrand factor than comparator products, thus minimizing circulating unbound FVIII and further reducing the potential risk of inhibitor development.
Collapse
|
88
|
Franchini M, Frattini F, Crestani S, Bonfanti C. Haemophilia B: current pharmacotherapy and future directions. Expert Opin Pharmacother 2012; 13:2053-63. [PMID: 22946758 DOI: 10.1517/14656566.2012.721780] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Hemophilia B is a rare hereditary hemorrhagic disorder characterized by deficiency of the clotting factor IX (FIX). Hemophilia B patients experience mild to severe bleeding complications according to the degree of FIX defect. Nowadays, the most challenging complication of individuals with hemophilia B is the development of alloantibodies, which render the standard replacement therapy with FIX concentrates ineffective, exposing them to a significantly increased morbidity and mortality. AREAS COVERED This review summarizes the most important events leading to the development of the current FIX products available for the treatment of hemophilia B patients. In addition, it focuses on the more recent advances in the production of new FIX molecules aimed at improving the clinical management of such patients. EXPERT OPINION Although the availability of plasma-derived FIX concentrates has greatly improved the clinical management of hemophilia B patients, the introduction of FIX products using recombinant DNA technology has represented the most significant therapeutic progress in hemophilia B therapy, ensuring an advanced level of safety. The development of rFIX products with extended half lives will further improve the therapeutic armamentarium for hemophilia B patients.
Collapse
Affiliation(s)
- Massimo Franchini
- Carlo Poma Hospital, Department of Transfusion Medicine and Hematology, Mantova, Italy.
| | | | | | | |
Collapse
|
89
|
Gilbert GE, Novakovic VA, Kaufman RJ, Miao H, Pipe SW. Conservative mutations in the C2 domains of factor VIII and factor V alter phospholipid binding and cofactor activity. Blood 2012; 120:1923-32. [PMID: 22613792 PMCID: PMC3433094 DOI: 10.1182/blood-2012-01-408245] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/01/2012] [Indexed: 11/20/2022] Open
Abstract
Factor VIII and factor V share structural homology and bind to phospholipid membranes via tandem, lectin-like C domains. Their respective C2 domains bind via 2 pairs of hydrophobic amino acids and an amphipathic cluster. In contrast, the factor V-like, homologous subunit (Pt-FV) of a prothrombin activator from Pseudonaja textilis venom is reported to function without membrane binding. We hypothesized that the distinct membrane-interactive amino acids of these proteins contribute to the differing membrane-dependent properties. We prepared mutants in which the C2 domain hydrophobic amino acid pairs were changed to the homologous residues of the other protein and a factor V mutant with 5 amino acids changed to those from Pt-FV (FV(MTTS/Y)). Factor VIII mutants were active on additional membrane sites and had altered apparent affinities for factor X. Some factor V mutants, including FV(MTTS/Y), had increased membrane interaction and apparent membrane-independent activity that was the result of phospholipid retained during purification. Phospholipid-free FV(MTTS/Y) showed increased activity, particularly a 10-fold increase in activity on membranes lacking phosphatidylserine. The reduced phosphatidylserine requirement correlated to increased activity on resting and stimulated platelets. We hypothesize that altered membrane binding contributes to toxicity of Pt-FV.
Collapse
Affiliation(s)
- Gary E Gilbert
- Department of Medicine, Veterans Administration Boston Healthcare System, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA.
| | | | | | | | | |
Collapse
|
90
|
Inaba H, Koyama T, Shinozawa K, Amano K, Fukutake K. Identification and characterization of an adenine to guanine transition within intron 10 of the factor VIII gene as a causative mutation in a patient with mild haemophilia A. Haemophilia 2012; 19:100-5. [PMID: 22776136 DOI: 10.1111/j.1365-2516.2012.02906.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2012] [Indexed: 11/28/2022]
Abstract
Haemophilia A is caused by various genetic mutations in the factor VIII gene (F8). However, after conventional analysis, no candidate mutation could be identified in the F8 of about 2% of haemophilia A patients. The F8 of a patient with mild congenital haemophilia A, in whom no candidate mutation was found in the exons or their flanking regions, was analysed in detail to identify the patient's aetiological genetic abnormality. We also characterized anti-FVIII antibody (inhibitor) development in this patient. Genomic DNA analysis revealed an adenine to guanine transition deep inside intron 10 (c.1478 + 325A>G) of F8 as a causative mutation. Analysis of the transcripts demonstrated that the majority of the patient's transcript was abnormal, with 226 bp of the intronic sequence inserted between exon 10 and 11. However, the analysis also indicated the existence of a small amount of normal transcript. Semi-quantification of ectopic F8 mRNA showed that about one-tenth of the normal mRNA level was present in the patient. After the use of a recombinant FVIII concentrate, the presence of an inhibitor was confirmed. The inhibitor was characterized as oligoclonal immunoglobulin IgG4 directed against both the A2 domain and light chain of the FVIII molecule with type I reaction kinetics of inhibition of FVIII activity. When no mutations are found by conventional analysis, deep intronic nucleotide substitutions may be responsible for mild haemophilia. The inhibitor development mechanism of the patient producing some normal FVIII was thought to be of interest.
Collapse
Affiliation(s)
- H Inaba
- Department of Laboratory Medicine, Tokyo Medical University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
91
|
LUSHER JM. Development and introduction of recombinant factor VIII - a clinician’s experience. Haemophilia 2012; 18:483-6. [DOI: 10.1111/j.1365-2516.2012.02804.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
92
|
The Relevance of Factor VIII (FVIII) Pharmacokinetics to TDM and Hemophilia A Treatment: Is B Domain–Deleted FVIII Equivalent to Full-Length FVIII? Ther Drug Monit 2012; 34:110-7. [DOI: 10.1097/ftd.0b013e318244fa5a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
93
|
Plantier JL, Saboulard D, Pellequer JL, Négrier C, Delcourt M. Functional mapping of the A2 domain from human factor VIII. Thromb Haemost 2012; 107:315-27. [PMID: 22234396 DOI: 10.1160/th11-07-0492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 11/24/2011] [Indexed: 11/05/2022]
Abstract
Coagulation factor VIII (FVIII) is a multidomain glycoprotein in which the FVIII A2 domain is a key structural element. We aimed at identifying residues within FVIII A2 domain that are crucial for the maintenance of the cofactor function. A high number (n=206) of mutants were generated by substituting original residues with alanine. The mutants were expressed in COS-1 cells and their antigen levels and procoagulant activities were measured. The residues were classified in three categories: those with a non-detrimental alteration of their activities (activity >50 % of control FVIII; n=98), those with a moderate alteration (15 %<activity<50%; n=45) and those that were severely affected (activity<15%; n=63). The mutants sensitive to mutation were retrieved in the HAMSTeRS database with a higher percentage than those that were not affected (58.8% vs. 9.2%). The results revealed the existence of clusters of residues that are sensitive (Arg418-Phe436, Thr459-Ile475, Ser535-Gly549, Asn618-Ala635) or not (Leu398-Arg418, Pro485-Asp500, Gly506-Gly520, Pro596-Asp605) to mutations. The stretches of residues sensitive to mutations were buried within the molecule suggesting that these amino acids participate in the maintenance of the A2 domain structure. In contrast, residues resistant to mutations formed external loops without well- defined structures suggesting that these loops were not crucial for the process of factor X activation. This study provided a detailed map of the FVIII A2 domain between residues 371 and 649, identifying residues crucial for maintaining FVIII function and residues that can be mutated without jeopardising the coagulant activity.
Collapse
Affiliation(s)
- Jean-Luc Plantier
- Laboratoire d’Hémobiologie EA4174-IFR62 Faculté de Médecine RTH Laennec, Université de Lyon, Lyon, France.
| | | | | | | | | |
Collapse
|
94
|
Greene TK, Lambert MP, Poncz M. Ectopic platelet-delivered factor (F) VIII for the treatment of Hemophilia A: Plasma and platelet FVIII, is it all the same? ACTA ACUST UNITED AC 2012; Suppl 1. [PMID: 24319630 DOI: 10.4172/2157-7412.s1-001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hemophilia A is the most common inherited bleeding diathesis and is due to a deficiency of functional coagulation factor (F) VIII. Most patients have a severe deficiency and require a program of prophylactic plus acute infusions of recombinant FVIII to prevent significant joint and other target organ damage. One of the greatest challenges remaining in the care of these patients is that one fifth to third of the patients develop inhibitors to the infused proteins. While a significant portion of such inhibitors can be either overcome or the inhibitors eliminated, some patients with persistent and significant titers of inhibitors need to rely on second tier therapies that are not as effective at preventing significant bleeding morbidity or mortality. A number of groups have been developing therapeutic strategies for FVIII gene therapy for this disorder. Virtually all of these therapies have in common a rise in the plasma level of FVIII, and interpretation of their efficacy is straightforward related to levels achieved. However, several groups have also shown that FVIII can be ectopically expressed in developing megakaryocytes, where although plasma FVIII levels remain undetectable, this FVIII can be released and be effective at sites of platelet activation. Moreover, it is clear that this platelet (p) FVIII is protected to a degree from inhibitors, making pFVIII a particularly attractive strategy for gene therapy for hemophilia A. Yet at the same time, we have shown that pFVIII has a different availability and distribution in a growing thrombus than plasma FVIII. The clinical implications and challenges of these findings as murine and canine hemophilia A preclinical studies go forward with pFVIII are discussed.
Collapse
Affiliation(s)
- Teshell K Greene
- Department of Pediatrics, The Children's Hospital of Philadelphia and the University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
95
|
Sharathkumar AA, Carcao M. Clinical advances in hemophilia management. Pediatr Blood Cancer 2011; 57:910-20. [PMID: 21755590 DOI: 10.1002/pbc.23193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 04/11/2011] [Indexed: 01/19/2023]
Abstract
Hemophilia is an excellent example in medicine where clinical translation of basic science discoveries has transformed the gloomy outlook of the disease. This review provides an overview of clinical advances in hemophilia management with a specific focus on the molecular heterogeneity of the disease and progress in management of patients with inhibitors. Novel therapeutics and the emerging ethical issues in the field of hemophilia are also discussed.
Collapse
Affiliation(s)
- Anjali A Sharathkumar
- Division of Hematology/Oncology and Stem Cell Transplantation, Department of Pediatrics, Children's Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60614-3363, USA.
| | | |
Collapse
|
96
|
DeAngelis JP, Varfaj F, Wakabayashi H, Fay PJ. The role of P4-P3' residues flanking Arg336 in facilitating activated protein C-catalyzed cleavage and inactivation of factor VIIIa. Thromb Res 2011; 128:470-6. [PMID: 21470668 PMCID: PMC3202615 DOI: 10.1016/j.thromres.2011.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 03/02/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Activated protein C (APC) inactivates factor VIIIa (FVIIIa) through cleavages at Arg336 in the A1 subunit and Arg562 in the A2 subunit. Proteolysis at Arg336 occurs 25-fold faster than at Arg562. Replacing residues flanking Arg336 en bloc with the corresponding residues surrounding Arg562 markedly reduced the rate of cleavage at Arg336, indicating a role for these residues in the catalysis mechanism. MATERIALS AND METHODS To assess the contributions of individual P4-P3' residues flanking the Arg336 site to cleavage efficiency, point mutations were made based upon those flanking Arg562 of FVIIIa (Pro333Val, Gln334Asp, Leu335Gln, Met337Gly, Lys338Asn, Asn339Gln) and selected residues flanking Arg506 of FVa (Leu335Arg, and Lys338Ile). APC-catalyzed inactivation of the FVIII variants and cleavage of FVIIIa subunits were monitored by FXa generation assays and Western blotting. RESULTS Specific activity values of the variants were 60-135% of the wild type (WT) value. APC-catalyzed rates of cleavage at Arg336 remained similar to WT for the Pro333Val and Lys338Ile variants and was modestly increased for the Asn339Gln variant; while rates were reduced ~2-3-fold for the Gln334Asp, Leu335Gln, Leu335Arg, and Lys338Asn variants, and 5-fold for the Met337Gly variant. Rates for cofactor inactivation paralleled cleavage at the A1 site. APC slowly cleaves Arg372 in FVIII, a site responsible for procofactor activation. Using FVIII as substrate for APC, the Met337Gly variant yielded significantly greater activation compared with WT FVIII. CONCLUSIONS These results show that individual P4-P3' residues surrounding Arg336 are in general more favorable to cleavage than those surrounding the Arg562 site.
Collapse
Affiliation(s)
- Jennifer P DeAngelis
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
97
|
Pipe SW, Miao H, Butler SP, Calcaterra J, Velander WH. Functional factor VIII made with von Willebrand factor at high levels in transgenic milk. J Thromb Haemost 2011; 9:2235-42. [PMID: 21920013 PMCID: PMC3444248 DOI: 10.1111/j.1538-7836.2011.04505.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Current manufacturing methods for recombinant human factor VIII (rFVIII) within mammalian cell cultures are inefficient, hampering the production of sufficient amounts for affordable, worldwide treatment of hemophilia A. However, rFVIII has been expressed at very high levels by the transgenic mammary glands of mice, rabbits, sheep, and pigs. Unfortunately, it is secreted into milk with low specific activity, owing in part to the labile, heterodimeric structure that results from furin processing of its B domain. OBJECTIVES To express biologically active rFVIII in the milk of transgenic mice through targeted bioengineering. METHODS Transgenic mice were made with a mammary-specific FVIII gene (226/N6) bioengineered for efficient expression and stability, encoding a protein containing a B domain with no furin cleavage sites. 226/N6 was expressed with and without von Willebrand factor (VWF). 226/N6 was evaluated by ELISA, SDS-PAGE, western blot, and one-stage and two-stage clotting assays. The hemostatic activity of immunoaffinity-enriched 226/N6 was studied in vivo by infusion into hemophilia A knockout mice. RESULTS AND CONCLUSIONS With or without coexpression of VWF, 226/N6 was secreted into milk as a biologically active single-chain molecule that retained high specific activity, similar to therapeutic-grade FVIII. 226/N6 had > 450-fold higher IU mL(-1) than previously reported in cell culture for rFVIII. 226/N6 exhibited similar binding to plasma-derived VWF as therapeutic-grade rFVIII, and intravenous infusion of transgenic 226/N6 corrected the bleeding phenotype of hemophilia A mice. This provides proof-of-principle for the study of expression of 226/N6 and perhaps other single-chain bioengineered rFVIIIs in the milk of transgenic livestock.
Collapse
Affiliation(s)
- Steven W. Pipe
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Hongzhi Miao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Stephen P. Butler
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA
| | - Jennifer Calcaterra
- Department of Chemical & Biomolecular Engineering, University of Nebraska, Lincoln, NE
| | - William H. Velander
- Department of Chemical & Biomolecular Engineering, University of Nebraska, Lincoln, NE
| |
Collapse
|
98
|
Importance of a factor VIIIc-like glycoprotein expressed in capillary endothelial cells (eFactor VIIIc) in angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 705:453-64. [PMID: 21618124 DOI: 10.1007/978-1-4419-7877-6_24] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
|
99
|
Hermans C, Brackmann HH, Schinco P, Auerswald G. The case for wider use of recombinant factor VIII concentrates. Crit Rev Oncol Hematol 2011; 83:11-20. [PMID: 21964312 DOI: 10.1016/j.critrevonc.2011.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 08/05/2011] [Indexed: 12/21/2022] Open
Abstract
The introduction of clotting factor concentrates led to major advances in hemophilia care. Rather than simply providing an alternative to plasma-derived concentrates, the introduction in the 1990s of recombinant concentrates added value to replacement therapy particularly with respect to prophylaxis and immune-tolerance induction. While the safety of plasma-derived concentrates has improved considerably, these concentrates may still pose an infectious risk through as-yet unknown pathogens and poor impurity constituent characterization. Recombinant concentrates are increasingly used because of their benefits in pathogen safety, convenience and the potential for unfettered supply. Yet worldwide they remain accessible only to a limited number of patients due to fear of the potential for inhibitor development, overestimation of their costs and underestimation of their benefits. This article reviews the characteristics and properties of recombinant FVIII concentrates to help physicians and patient representatives promote the right of access of patients to the safest products.
Collapse
Affiliation(s)
- Cedric Hermans
- Hemostasis and Thrombosis Unit, Haemophilia Clinic, CliniquesUniversitaires Saint-Luc, Catholic University of Louvain, 1200 Brussels, Belgium.
| | | | | | | |
Collapse
|
100
|
Abstract
Haemophilia A and B are X-linked bleeding disorders due to the inherited deficiency of factor VIII or factor IX, respectively. Of the approximately 1 per 5000-10000 male births affected by haemophilia, 80% are deficient in factor VIII and 20% are deficient in factor IX. Haemophilia is characterized by spontaneous and provoked joint, muscle, gastrointestinal and CNS bleeding leading to major morbidity and even mortality if left untreated or under-treated. The evolution of haemophilia management has been marked by tragedy and triumph over recent decades. Clotting factors and replacement strategies continue to evolve for patients without inhibitors. For patients with an inhibitor, factor replacement for acute bleeding episodes and immune tolerance, immune modulation and extracorporeal methods for inhibitor reduction are the cornerstone of care. In addition, adjuvant therapies such as desmopressin, antifibrinolytics and topical agents also contribute to improved outcomes for patients with and without inhibitors. The future direction of haemophilia care is promising with new longer-acting clotting factors and genetic therapies, including gene transfer and premature termination codon suppressors. With these current and future treatment modalities, the morbidity and mortality rates in patients with haemophilia certainly will continue to improve.
Collapse
Affiliation(s)
- Trisha Wong
- Puget Sound Blood Center, Seattle, Washington, USA
| | | |
Collapse
|