THE PAST TWO DECADES have brought remarkable progress in our understanding of the molecular basis of hemophilia A. This disease, which has already been documented as a familial bleeding tendency in the fifth century,1 still persists as the most common hemorrhagic disorder, affecting 1 in approximately 5,000 males.2 Hemophilia has been associated with deficiency of a plasma component since 1937, when Patek and Taylor3 showed that the clotting defect of hemophilic plasma could be corrected by plasma of a normal individual. This component was called “antihemophilic factor,” or “factor VIII” according to the more recent nomenclature. Subsequent studies using preparations enriched in factor VIII activity have established factor VIII as being the cofactor of activated factor IX in the factor X–activating complex of the intrinsic coagulation pathway.4 However, the molecular entity of factor VIII has remained unidentified until the early 1980s, when the protein was purified to complete homogeneity, and its cDNA was cloned.5-8 This breakthrough has triggered numerous studies on the genetic and molecular basis of hemophilia A, and consequently our knowledge on the structure and function of the factor VIII protein has been rapidly expanding since then.

The present review focuses on the “life cycle” of factor VIII, which comprises the sequence of events between biosynthesis and clearance of the protein (see Fig 1). These processes are discussed in view of our current knowledge on factor VIII structure and function, with particular reference to the proteolytic modulation of factor VIII, and its assembly into the factor X activating complex.

Fig. 1.

The lifespan of factor VIII. Factor VIII is synthesized by various tissues, including liver, kidney, and spleen, as an inactive single-chain protein. After extensive posttranslational processing, factor VIII is released into the circulation as a set of heterodimeric proteins. This heterogenous population of factor VIII molecules readily interacts with vWF, which is produced and secreted by vascular endothelial cells. Upon triggering of the coagulation cascade and subsequent generation of serine proteases, factor VIII is subject to multiple proteolytic cleavages. These cleavages are associated with dramatic changes of the molecular properties of factor VIII, including dissociation of vWF and development of biological activity. After conversion into its active conformation, and participation in the factor X activating complex, activated factor VIII rapidly looses its activity. This process is governed by both enzymatic degradation and subunit dissociation.

Fig. 1.

The lifespan of factor VIII. Factor VIII is synthesized by various tissues, including liver, kidney, and spleen, as an inactive single-chain protein. After extensive posttranslational processing, factor VIII is released into the circulation as a set of heterodimeric proteins. This heterogenous population of factor VIII molecules readily interacts with vWF, which is produced and secreted by vascular endothelial cells. Upon triggering of the coagulation cascade and subsequent generation of serine proteases, factor VIII is subject to multiple proteolytic cleavages. These cleavages are associated with dramatic changes of the molecular properties of factor VIII, including dissociation of vWF and development of biological activity. After conversion into its active conformation, and participation in the factor X activating complex, activated factor VIII rapidly looses its activity. This process is governed by both enzymatic degradation and subunit dissociation.

Close modal
Factor VIII gene.

The gene of factor VIII is located at the tip of the long arm of the X chromosome.5 It spans over 180 kb, and as such is one of the largest genes known. Its transcription may require several hours assuming a transcription rate of 10 nucleotides per second, and yields a 9-kb mRNA product.5 The factor VIII gene comprises 26 exons, which encode a polypeptide chain of 2351 amino acids.6-8 This includes a signal peptide of 19 and a mature protein of 2332 amino acids. Analysis of the deduced primary structure determined from the cloned factor VIII cDNA showed the presence of a discrete domain structure: A1-a1-A2-a2-B-a3-A3-C1-C26-8 (Fig2). The A domains display approximately 30% homology to each other. These domains further display a similar extent of homology to the copper-binding protein ceruloplasmin and to factor V, the cofactor in the prothrombinase complex.9 The A domains are bordered by short spacers (a1, a2, and a3) that contain clusters of Asp and Glu residues, the so-called acidic regions. The C domains are structurally related to the C domains of factor V. In addition, the lipid-binding lectin discoidin I, human and murine milk fat globule proteins, and a putative neuronal cell adhesion molecule from Xenopus laevis share amino acid sequence similarity to the factor VIII C domains.10-12 The B domain is unique in that it exhibits no significant homology with any other known protein.

Biosynthesis of factor VIII.

Several tissues have the potential of expressing the factor VIII gene. Factor VIII mRNA has been demonstrated in a variety of tissues, including spleen, lymph nodes, liver, and kidney.13-15Transplantation studies in hemophilic animals showed that organs such as lung and spleen indeed contribute to the presence of circulating factor VIII.16,17 However, the liver most likely provides the primary source of factor VIII. This view is supported by liver perfusion18,19 and by liver transplantation studies in both animals and humans.20-22 A number of cases concerning hemophilic patients have been reported in which transplantation resulted in sustained, normalized levels of factor VIII.21 22 

Several lines of evidence indicate that, within the liver, hepatocytes are the major factor VIII–producing cells. First, factor VIII mRNA is present in hepatocytes but not in sinusoidal cells.13Second, the promotor region of the factor VIII gene comprises responsive elements that are characteristic for hepatocyte-specific expression.23 Finally, in immuno-ultrastructural studies factor VIII protein was detected in the rough endoplasmatic reticulum and the Golgi apparatus of hepatocytes.24 It should be mentioned that other reports showed the presence of factor VIII in hepatic endothelium using histochemical techniques.25-27This is unexpected because these cells appear to lack factor VIII mRNA. It is possible that the latter observation reflects surface binding of factor VIII or internalization rather than factor VIII biosynthesis.

Secretion of factor VIII.

Studies on factor VIII biosynthesis and secretion have been limited by the lack of human cell lines that properly express significant amounts of factor VIII. Analysis of the factor VIII secretion process has therefore been restricted to autologous gene expression.28These studies showed that, in general, factor VIII is poorly expressed. Low expression is associated with a low level of steady-state mRNA29 and inefficient secretion.30 The initial stage of secretion involves the translocation of the mature 2332 amino acid polypeptide into the lumen of the endoplasmatic reticulum (ER), where N-linked glycosylation occurs. Within the ER, factor VIII appears to interact with a number of chaperone proteins, including calreticulin, calnexin, and the Ig-binding protein (BiP).31-34 Due to the interaction with these chaperone proteins, a significant proportion of the factor VIII molecules is retained within the ER, thereby limiting the transport of factor VIII to the Golgi apparatus. The mechanism reponsible for the transport from the ER to the Golgi apparatus is not elucidated yet. However, recent studies indicate that this step involves an intracellular membrane lectin: endoplasmatic reticulum-Golgi intermediate compartment-53 (ERGIC-53).35 

Within the Golgi apparatus, factor VIII is subject to further processing, including modification of the N-linked oligosaccharides to complex-type structures, O-linked glycosylation, and sulfation of specific Tyr-residues (Fig 2). In addition, factor VIII is among the many proteins that undergoes intracelullar proteolysis.36-39 The middle part and the carboxyterminal region of the B domain comprise a motif (Arg-X-X-Arg), which is similar to the motif that is recognized by intracellular proteases of the subtilisin-like family.38 39 The responsible endoprotease mediating intracellular factor VIII proteolysis, however, has not been identified. The Arg-X-X-Arg motif allows proteolysis at Arg1313 and at Arg1648. The latter event disrupts the covalent linkage of the factor VIII heavy chain (A1-a1-A2-a2-B) and light chain (a3-A3-C1-C2), giving rise to the heterodimeric molecule that circulates in plasma.

The factor VIII heavy and light chain remain noncovalently associated through the A1 and A3 domain in a metal-ion–dependent manner.7,40-42 Considering the structural homology of factor VIII to the copper-binding protein ceruloplasmin, it is not surprising that copper ions have been found in factor VIII as well.43,44 One molecule of copper is present per molecule of factor VIII. Most likely, the copper-ion binding site is composed of residues His265, Cys310, His315, and Met320 within the A1 domain.44 Binding of copper ions to this site may allow the A1 domain to adopt an A3-domain binding conformation. Alternatively, copper ions may directly bridge the A1 and A3 domain by interacting with both domains simultaneously. Whether only copper ions are involved in the association between heavy and light chain is unclear. Whereas in the absence of other metal ions copper ions are ineffective in promoting reassembly of dissociated heavy and light chain, calcium or manganese ions are considerably more efficient in this respect.45-47a However, specific activity of factor VIII dimers that were reassociated in the presence of calcium ions, is markedly enhanced upon the addition of copper ions.47a Apparently, copper ions serve an auxiliary role to enhance cofactor function of factor VIII. These observations suggest that multiple sites may be involved in the association between heavy and light chain. Irrespective of the precise mechanism, it is clear that metal ions serve an important role in maintaining the heterodimeric structure of secreted factor VIII.

Factor VIII secretion and hemophilia A.

Aberrant biosynthesis or secretion may result from several defects. Obviously, gross deletions or rearrangements may result in impaired transcription, RNA processing, or translation. No data are reported on the secretion of such gene products or on the stability within the circulation provided that these gene products are actually secreted. Defective secretion may further be caused by apparently minor gene defects like single missense mutations. Frequently known missense mutations associated with low levels of factor VIII are located in codon 2307, resulting in replacement of Arg2307 by Gln or Leu.48,49 Both mutations have been analyzed using recombinant factor VIII mutants expressed in mouse fibroblasts50 and COS-1 monkey kidney cells.51Both proteins appear to be functionally normal, but are poorly secreted. The majority of the retained mutants are targeted into an ER-associated degradation pathway. The mechanism responsible for intracellular retention of these mutated factor VIII molecules is unknown.

Reduced levels of factor VIII protein may also result from defects located outside the factor VIII gene. One striking example concerns patients having combined factor V and VIII deficiency. The gene responsible for combined factors V and VIII deficiency has been mapped to the long arm of chromosome 18, between markers D18S849 and D18S1103,52,53 whereas the genes for factors V and VIII are located at chromosomes 1 and X, respectively. Recently, the unknown gene has been idenfied to encode the intracellular membrane lectin ERGIC-53, a resident protein of the ER-Golgi intermediate compartment.35 Indeed, affected individuals displayed mutations in this gene, in association with a complete lack of expression of ERGIC-53.35 Apparently, ERGIC-53 contributes to the secretion process of factors V and VIII, presumably by acting as a chaperone protein. Identification of the underlying mechanism should provide more insight in the intracellular routing and secretion of both cofactors.

Binding sites for vWF.

Immediately after its release into the circulation, the factor VIII heterodimer interacts with its carrier protein vWF to form a tight, noncovalent complex. Each monomer of the multimeric vWF protein is able to bind one factor VIII molecule with high affinity (kd < 0.5 nmol/L).54-57 In vivo, however, the stoichiometry is limited by the number of factor VIII molecules present, resulting in approximately a 1:50 ratio.

Two peptide regions of factor VIII are implicated to be involved in binding vWF: one at the aminoterminal end of intact factor VIII light chain,55-59 and one at the carboxyterminal end (residues 2303-2332).57,60-62 Using proteolytically derived fragments of factor VIII light chain, it was shown that both these individual regions are capable of binding vWF.57 However, the affinity of these fragments for vWF is markedly lower compared with the intact factor VIII heterodimer (two and three orders of magnitude for the aminoterminal and carboxyterminal end, respectively).57Apparently, both ends of factor VIII light chain act synergistically in the binding of vWF.

With respect to vWF binding to the aminoterminal region of factor VIII light chain, it appears that residues 1649 to 1671, thus including sulfated residue Tyr1664, are dispensable for vWF binding.57,63,64 In addition, the Arg1689-Ser1690 cleavage site has to be intact,57 suggesting that residues carboxyterminal of this thrombin cleavage site contribute to binding as well. Recombinant factor VIII synthesized in the presence of an inhibitor of sulfation displays reduced binding to vWF,63,65 suggesting a role for sulfated Tyr1680 in the interaction with the carier protein. Indeed, replacement of Tyr1680 by Phe results in loss of high-affinity binding to vWF, allowing only a low-affinity interaction.63,65 66 The presence of Tyr1680 in its sulfated form thus contributes to optimal binding of factor VIII to vWF.

Factor VIII interactions modulated by vWF.

One functional aspect of factor VIII-vWF complex formation may be to prevent premature binding of factor VIII to components of the factor X–activating complex. For instance, binding of factor VIII light chain to factor IXa is inhibited by vWF.67 Because the affinity of factor VIII for vWF exceeds that for factor IXa by approximately 100-fold,56 67 factor VIII highly favors vWF binding over factor IXa binding. The mechanism by which vWF inhibits factor IXa binding is not yet elucidated. Because binding of both proteins by factor VIII light chain seems to involve distinct parts of the molecule, it is unlikely that vWF competes with factor IXa for binding to the same site. Mechanisms that could contribute to inhibition include sterical hindrance and alteration of the factor VIII conformation so that factor IXa cannot bind.

Whereas factor IXa and vWF bind at different sites, vWF and phospholipids both bind to the C2-domain region 2303-2332.62,68 Using a human anti–factor VIII antibody, it has been shown that these sites, although in very close proximity, do not completely overlap.69 Nevertheless, the close proximity of these sites may explain the observations that binding of factor VIII to vWF is incompatible with factor VIII binding to membrane surfaces.70-73 It should be noted that, in comparison with noncomplexed factor VIII, factor VIII in complex with vWF is less susceptible to proteolytic attack by various lipid-binding proteases.74-77 These vitamin K–dependent serine proteases, which include activated protein C (APC) and factor Xa, require to assemble with factor VIII at a membrane surface for efficient proteolysis of factor VIII.76,78 In contrast, thrombin displays proteolytic activity independent of a membrane surface. Indeed, vWF does not protect factor VIII against cleavage by thrombin.79-81 Cleavage of factor VIII by thrombin results in loss of vWF binding and conversion of factor VIII into its active conformation.

vWF and hemophilia A.

The association with vWF serves an important role in factor VIII physiology, as vWF functions as a stabilizer of the heterodimeric structure of factor VIII.45,82-84 The physiological relevance of complex formation is particularly apparent in patients with von Willebrand disease (vWD) (type 3), who have no detectable vWF protein. Not only do these patients have a secondary deficiency of factor VIII, but they also have a considerably reduced half-life of intravenously administred factor VIII.83,85-88 This phenotype is also observed in patients with vWD with the so-called Normandy variant, which is defined as type 2N. Despite normal levels of circulating vWF, factor VIII levels are severely reduced.89 90 These patients harbor a mutation in the factor VIII–binding domain of vWF, which results in defective binding to vWF.

With regard to factor VIII, two distinct basepair substitutions have been reported that are associated with impaired complex assembly.49,91 92 Both mutations result in the replacement of one single amino acid, Tyr1680, and are associated with hemophilia A. So far, no mutations located within the C2 domain region have been reported that are associated with reduced affinity for vWF.

Cleavage sites associated with factor VIII activation.

Within the factor X–activating complex, the proteolytic activity of factor IXa is markedly enhanced by factor VIII. It has been well established that proteolysis of factor VIII is required for the generation of its cofactor activity.79,93-97 The uncleaved factor VIII procofactor lacks the ability to enhance factor IXa activity.47,98 Enzymes that are able to endow factor VIII with its cofactor activity include thrombin and factor Xa.79,93,98-102 Thrombin cleaves factor VIII at one specific site within the light chain, Arg1689, and at two sites in the heavy chain: Arg372 and Arg740(Fig 3).79Proteolysis of factor VIII heavy chain by factor Xa involves three sites: Arg336, and the two thrombin-cleavage sites Arg372 and Arg740(Fig 3).79 With regard to factor VIII light chain, factor Xa is able to cleave at Arg1689, a site that is shared with thrombin, but also at Arg1721, a site that is specific for factor Xa.79 It should be noted that this site is cleaved in human factor VIII, but not in porcine factor VIII.103 It has been unclear whether cleavage at Arg1721 contributes to factor VIII activation or inactivation, because prolonged incubation with factor Xa results in loss of factor VIII activity in parallel with cleavages at positions 336 and 1721.79 This has been resolved by studying reassociated dimers of intact factor VIII heavy chain with either thrombin– or factor Xa–cleaved factor VIII light chain.47 The resulting factor VIII dimers were functionally indistinguishable, demonstrating that factor Xa–cleavage of the light chain is not associated with inactivation. It has been reported that factor Xa–activated factor VIII displays less activity than thrombin-activated factor VIII.79,98 102 Because cleavage of the light chain cannot be responsible for this phenomenon, this is most likely due to additional factor Xa–cleavage at Arg336in the factor VIII heavy chain.

Fig. 2.

The factor VIII protein. Mature factor VIII consists of 2332 amino acids, which are arranged in a discrete domain structure: A1 (residues 1-336), A2 (373-710), B (741-1648), A3 (1690-2019), C1 (2020-2172), and C2 (2173-2332). The A domains are bordered by acidic regions a1 (337-372), a2 (711-740), and a3(1649-1689). Disulfide Bridges: Using B-domainless factor VIII, seven disulfide bonds have been identified: residues 153 and 179, 248 and 329 (A1 domain), 528 and 554, 630 and 711 (A2 domain), 1832 and 1858, 1899 and 1903 (A3 domain), and 2021 and 2169 (C1 domain).196Within the C2 domain, residues 2174 and 2326 most likely also form a disulfide bridge. Free cysteine-residues have been identified at positions 310, 692, and 2000.196 Cys528 and Cys1858 may be present as free cysteines, because these residues are reactive toward a sulfhydryl-specific fluorphor.197 With regard to the Cys-residues in the B-domain it is unknown whether they are free or linked.N-Linked Glycosylation: Factor VIII contains 25 consensus sequences (Asn-Xxx-Thr/Ser) that allow N-linked glycosylation. Using either full-length or B-domainless factor VIII, the majority of these sites have been shown to be glycosylated: residues 42 and 239 (A1 domain), residues 757, 784, 828, 900, 963, 1001, 1005, 1055, 1066, 1185, 1255, 1259, 1282, 1300, 1412, and 1442 (B domain), residue 1810 (A3 domain), and residue 2118 (C2 domain).198-200Nonglycosylated residues are present at positions 943 and 1384 (B domain) and at position 1685 (a3 acidic region). Residue 582 (A2 domain) has been reported to be nonglycosylated in two studies,199,200 whereas one study reported this residue to be partially glycosylated.198 Finally, it remains to be investigated whether residue 1512 (B domain) is glycosylated. Tyrosine Sulfation: The acidic regions contain consensus sequences that allow sulfation of Tyr-residues at positions 346 (a1 region), 718, 719, 723 (a2 region), 1664, and 1680 (a3 region). Analysis using recombinant proteins established that all sites indeed can be sulfated.

Fig. 2.

The factor VIII protein. Mature factor VIII consists of 2332 amino acids, which are arranged in a discrete domain structure: A1 (residues 1-336), A2 (373-710), B (741-1648), A3 (1690-2019), C1 (2020-2172), and C2 (2173-2332). The A domains are bordered by acidic regions a1 (337-372), a2 (711-740), and a3(1649-1689). Disulfide Bridges: Using B-domainless factor VIII, seven disulfide bonds have been identified: residues 153 and 179, 248 and 329 (A1 domain), 528 and 554, 630 and 711 (A2 domain), 1832 and 1858, 1899 and 1903 (A3 domain), and 2021 and 2169 (C1 domain).196Within the C2 domain, residues 2174 and 2326 most likely also form a disulfide bridge. Free cysteine-residues have been identified at positions 310, 692, and 2000.196 Cys528 and Cys1858 may be present as free cysteines, because these residues are reactive toward a sulfhydryl-specific fluorphor.197 With regard to the Cys-residues in the B-domain it is unknown whether they are free or linked.N-Linked Glycosylation: Factor VIII contains 25 consensus sequences (Asn-Xxx-Thr/Ser) that allow N-linked glycosylation. Using either full-length or B-domainless factor VIII, the majority of these sites have been shown to be glycosylated: residues 42 and 239 (A1 domain), residues 757, 784, 828, 900, 963, 1001, 1005, 1055, 1066, 1185, 1255, 1259, 1282, 1300, 1412, and 1442 (B domain), residue 1810 (A3 domain), and residue 2118 (C2 domain).198-200Nonglycosylated residues are present at positions 943 and 1384 (B domain) and at position 1685 (a3 acidic region). Residue 582 (A2 domain) has been reported to be nonglycosylated in two studies,199,200 whereas one study reported this residue to be partially glycosylated.198 Finally, it remains to be investigated whether residue 1512 (B domain) is glycosylated. Tyrosine Sulfation: The acidic regions contain consensus sequences that allow sulfation of Tyr-residues at positions 346 (a1 region), 718, 719, 723 (a2 region), 1664, and 1680 (a3 region). Analysis using recombinant proteins established that all sites indeed can be sulfated.

Close modal
Fig. 3.

Limited proteolysis of factor VIII. The major part of factor VIII circulates as a set of heterogenous dimers, consisting of a light (a3-A3-C1-C2) and heavy chain (A1-a1-A2-a2-B). The heavy chain is variably sized due to limited proteolysis within the B domain. Some of these cleavages may occur intracellularly at positions 1313 and 1648 (open downward arrows). Factor VIII can be converted into its active form by proteolysis in both the heavy and light chain by various serine proteases (closed downward arrows), including thrombin and factor Xa. Because proteolysis by factor Xa but not thrombin is inhibited by vWF, thrombin is probably the physiological activator of factor VIII. Proteolytic degradation of factor VIIIa proceeds through cleavages within the A1 and A2 domains by various serine proteases (upward arrows), and results in release of the a1 acidic region and bisecting of the A2 domain. In contrast to what has previously been assumed, cleavages within the light chain by factor IXa or factor Xa do not result in inactivation of factor VIII, but contribute to the development of factor VIII cofactor activity.

Fig. 3.

Limited proteolysis of factor VIII. The major part of factor VIII circulates as a set of heterogenous dimers, consisting of a light (a3-A3-C1-C2) and heavy chain (A1-a1-A2-a2-B). The heavy chain is variably sized due to limited proteolysis within the B domain. Some of these cleavages may occur intracellularly at positions 1313 and 1648 (open downward arrows). Factor VIII can be converted into its active form by proteolysis in both the heavy and light chain by various serine proteases (closed downward arrows), including thrombin and factor Xa. Because proteolysis by factor Xa but not thrombin is inhibited by vWF, thrombin is probably the physiological activator of factor VIII. Proteolytic degradation of factor VIIIa proceeds through cleavages within the A1 and A2 domains by various serine proteases (upward arrows), and results in release of the a1 acidic region and bisecting of the A2 domain. In contrast to what has previously been assumed, cleavages within the light chain by factor IXa or factor Xa do not result in inactivation of factor VIII, but contribute to the development of factor VIII cofactor activity.

Close modal

Because activation of factor VIII involves proteolysis of both its heavy and light chain, it is of interest to compare the relative contribution of each cleavage to the development of factor VIII cofactor function. The contribution of cleavage at Arg740to factor VIII activation is limited, because mutations at this position do not interfere with factor VIII activation or function.97 Selective cleavage of factor VIII at Arg372 by a snake venom–derived protease generates a factor VIII molecule that displays 60% of the activity of fully activated factor VIII.80 Factor VIII dimers cleaved within the light chain only have 25% to 30% of the activity displayed by fully activated factor VIII.47,104 These findings allow the conclusion that both cleavage at Arg372 and Arg1689 are required to exert full cofactor activity. This view is supported by the observation that recombinant factor VIII mutants containing replacements at either Arg372 or Arg1689 are unable to correct the clotting time of factor VIII–deficient plasma.97 

Acidic regions.

With regard to the cleavage sites at positions 372, 740, and 1689, it is noteworthy that these residues are located at the carboxyterminal end of an acidic sequence interconnecting adjacent domains. These acidic regions (a1, a2, and a3, see Fig 2) contain several Tyr residues in a sequence that meets the consensus features for tyrosine sulfation.105,106 Sulfated Tyr residues may serve a role in various processes, including protein-protein interactions.106 Because thrombin is known to interact with a variety of acidic, sulfated sequences, it has been proposed that acidic regions within factor VIII serve a role in thrombin activation of factor VIII.107 Replacement of Tyr346 (a1 region) or Tyr1664(a2 region) indeed results in factor VIII mutants that are activated by thrombin less efficiently.66 It should be mentioned that deletion of Tyr1664 leaves the activation kinetics of factor VIII by thrombin unaffected.108 109 

Replacement of Tyr-residues by Phe in the a2 acidic region (residues 718, 719, and 723) did not affect thrombin activation, but rather resulted in mutants that displayed reduced ability to stimulate factor IXa enzymatic activity.66 In contrast, recombinant factor VIII variants (both full-length and B-domainless) containing nonsulfated Tyr at these positions are activated normally and display full cofactor activity.110,111 Although both approaches reveal apparently contrasting findings with regard to the effect of Tyr-sulfation in the a2 region on factor VIII cofactor function, they allow the conclusion that sulfation of these Tyr-residues does not contribute to activation of factor VIII. However, the acidic nature of this a2 sequence appears to be of importance for activation of factor VIII. First, a monoclonal antibody directed against a2 inhibits thrombin activation of factor VIII, but does not interfere with factor VIII cofactor activity.112,113 Secondly, deletion of a2 or part thereof in B-domainless factor VIII results in molecules that require higher thrombin concentrations than normal factor VIII for efficient activation, but display normal factor IXa cofactor activity.110,112 Interestingly, in both cases it was observed that reduced thrombin activation was caused by a reduced cleavage efficiency at Arg372 and Arg1689, suggesting that the a2 acidic region influences cleavage in remote regions in the factor VIII molecule. This hypothesis is supported by observations using a factor VIII chimera with a replacement in the a2 region. In this chimeric molecule, residues 716-736 of a2 have been replaced by a sequence that is known to have a high affinity for thrombin, ie, the amino acid sequence 51-80 of heparin cofactor II.114 This chimeric protein proved more potent than normal factor VIII in correcting the clotting time of factor VIII–deficient plasma. The increased intrinsic activity is caused by an increased rate of thrombin cleavage at Arg372 and Arg1689 compared with normal factor VIII. Thus, these findings suggest that the a2 region promotes proteolytic activation of factor VIII.

Apart from their role in thrombin activation, the acidic regionsa1 and a3 contribute to factor VIII function also in an additional manner. The a1 region has been described to be involved in maintaining the stability of the factor VIII heterotrimer115 and in binding of factor X116,117 (see Factor VIII Inactivation section). Thea3 region is important for high affinity binding of vWF (see Assembly of the Factor VIII-vWF Complex section). Further, factor VIII heterodimers consisting of uncleaved heavy and light chain do not posses any cofactor activity.47,48 In contrast, factor VIII exclusively cleaved at position 1689, thus lacking the a3region, displays significant cofactor activity (approximately 25% of fully activated factor VIII).47 104 Apparently, the acidic region a3 functions as an activation peptide that needs to be cleaved off for exposure of cofactor activity.

Defective activation and hemophilia A.

Mutations resulting in replacement of amino acids at the factor VIII activation sites should predispose to hemorraghic diathesis. Indeed, missense mutations at Arg372, Ser373, and Arg1689 are associated with hemophilia A.49,118-124 With regard to the Arg1689mutation, biochemical data predict residual activity to be at least half of that of normal factor VIII, as cleavage at Arg372accounts for approximately 60% of total activity.80However, residual activity appears to range between <1% and ≈12%, thus lower than expected.49 Several possibilities may be considered to explain this apparent discrepancy. First, due to substitution of Arg1689, the vWF binding site including sulfated Tyr1680 is not cleaved from the light chain. Therefore, the factor VIII–vWF complex may fail to dissociate upon thrombin treatment. As a consequence, factor VIII is unable to interact with factor IXa and phospholipids, and thus cannot assemble into the factor X–activating complex (see below). Secondly, in most of the documented patients with a mutation at Arg1689, this residue is replaced by a Cys.49 This Cys-residue has the potential to form an extra disulfide bridge within the factor VIII light chain.125 126 It seems conceivable that this results in misfolding within the factor VIII light chain, which precludes biological activity. Finally, residual activity may be affected by the type of amino acid substitution. This view is supported by the notion that a similar discrepancy exist for substitutions at Arg372. For instance, Arg372 to Pro substitutions exclusively result in severe hemophilia A, whereas Arg372 to His substitutions result in a mild to moderate phenotype.

Regulation of complex assembly.

To activate factor X, factor IXa and factor VIIIa assemble into a membrane-bound complex. To maintain the hemostatic balance, this complex should only assemble after initiation of the coagulation cascade, implying that participation of factor VIII in this complex is subject to a delicate regulatory mechanism. In this regard, vWF plays a central role. Although factor IXa displays similar affinity for nonactivated and activated factor VIII,127 the factor IXa binding site within nonactivated factor VIII is unlikely to be accesible when factor VIII is in complex with vWF.67Furthermore, binding of factor VIII to the membrane surface is inhibited by vWF,70-73 suggesting that only activated factor VIII that is dissociated from vWF is able to bind to the membrane surface. However, it should be mentioned that the affinity of factor VIII for the membrane surface is dependent on the membrane composition, as affinities have been reported that differ 10- to 100-fold (10−9 to 10−11mol/L).72,128-131 Thus, under particular conditions the affinity of vWF and the membrane surface for factor VIII is similar. Apparently, a delicate balance may exist between factor VIII being in complex with vWF or the membrane surface. Because in direct binding studies vWF prevents binding of factor VIII to the membrane surface,70-73 a minor part of the factor VIII population probably is in complex with the membrane surface. This situation changes dramatically upon cleavage of factor VIII light chain, which results in a 1,600-fold decrease in affinity for vWF.57Because of this event, the balance will readily shift toward factor VIIIa binding to the membrane surface. This subsequently favors binding of factor IXa to factor VIIIa, which is no longer associated with vWF. Ultimately, this leads to the assembly of the membrane-bound factor VIIIa–factor IXa complex that activates factor X.

The role of the membrane surface in complex assembly.

The notion that in the absence of a membrane surface the generation of factor Xa by the factor VIIIa–factor IXa complex is negligible132 underscores the essential role of the membrane surface in the factor X–activating complex. The membrane surface may act in two distinct ways: first by positioning the enzyme-cofactor complex into an active conformation or, alternatively, by locating the enzyme and cofactor at the same site. At present, data have been reported that are in support of both mechanisms. On the one hand, it has been shown that the affinity of factor IXa for factor VIIIa is increased 2,000-fold in the presence of phospholipids,133 which suggests that the second mechanism is dominant. On the other hand, the affinity of (activated) factor VIII for factor IXa is reported to be similar in the presence (kd = 10−8 to 10−9mol/L)104,127,132,134,135 and absence (kd = 10−8 mol/L)67,132 of a phospholipid surface. Gilbert and Arena132 showed that in the presence of phospholipids the catalytic activity of the enzyme-cofactor complex is increased 1,500-fold. These data favor the view that the membrane surface positions the enzyme and cofactor in a conformation that allows efficient substrate cleavage.

Location of factor IXa interactive sites.

The interaction between factor VIIIa and factor IXa has been investigated in several elegant studies using factor IXa molecules that carry a fluorescent-label in the active site.127,134-139 It became evident that in the presence of phospholipids, factor VIIIa induces a conformational change in the factor IXa protease domain. In addition, maximal changes in the factor IXa protease domain require the presence of the factor VIIIa A2-domain,134 suggesting that the A2 domain contains a factor IXa interactive site. Indeed, studies using a series of synthetic peptides showed that factor IXa binding can be attributed to the A2-domain sequence 558 to 565.137Furthermore, a region within the carboxyterminal part of the A2 domain (residues 698 to 710) also has been proposed to comprise a factor IXa–binding site.140 141 

Besides factor VIII heavy chain, the light chain also contributes to factor IXa binding. In equilibrium binding studies, isolated factor VIII light chain proved to bind factor IXa with high affinity.67 Moreover, factor VIII light chain and the intact factor VIII heterodimer are indistinguishable in terms of affinity for factor IXa, indicating that high-affinity binding to factor IXa is mediated by the factor VIII light chain. Binding of factor VIII light chain to factor IXa was found to be inhibited by the A3-domain directed monoclonal antibody CLB-CAg A, a strong inhibitor of factor VIII activity.67,113,142 By using synthetic peptides, the A3-domain sequence 1811 to 1818 has been identified as a site that binds factor IXa.142 Thus, interaction with factor IXa involves at least three sites on factor VIII: residues 558-565, 698-710, and 1811-1818.

Three-dimensional model of factor VIII and factor IXa.

Three-dimensional representations of the enzyme and cofactor have been published, based on factor IXa crystallography143 and factor VIII homology modeling144,145(Fig 4). So far, it is unknown which residues in the factor IXa molecule are involved in binding factor VIII. However, the location of these sites should fit with the location of their counterparts on the factor VIII molecule. Although the amino acid numbering suggests that the factor IXa–binding regions are located in completely different parts of the factor VIII protein, the three-dimensional model indicates that the factor IXa–binding sites are in close vicinity, and are exposed at the same side of the molecule (Fig 4). Matching of the factor VIII and factor IXa models suggests that factor IXa comprises distinct sites involved in factor VIIIa binding, interacting with the A2 or A3 domain. Factor VIIIa binding appears to be mediated by both the factor IXa light chain and protease domain. The involvement of the protease domain is in agreement with the observation by Bajaj et al146,147 that the stimulation of factor IXa proteolytic activity by factor VIIIa is inhibited by a monoclonal antibody directed against the protease domain residues 231-265. This suggests that this protease domain region comprises a factor VIII–binding site. However, recombinant factor IXa molecules in which residues in the antibody-binding epitope have been mutated combine a strongly reduced affinity for the antibody with normal biological activity.148 Because normal activity is associated with normal factor VIII binding, these findings leave the exact location of the factor VIII binding site in the factor IXa heavy chain unidentified. Irrespective of its precise location, this site on the factor IXa heavy chain presumably interacts with the factor VIII A2 domain, because this domain induces the largest change in the conformation of the factor IXa active site.134 

Fig. 4.

Model of the factor VIII and factor IXa molecules. Shown are representiations of porcine factor IXa (Protein Data Bank accession code 1pfx) and the triplicated A-domains of human factor VIII (Hemophilia A web site, http://europium.mrc.rpms.ac.uk), which are derived from crystallography and homology modeling, respectively. Factor IXa binding region in the factor VIII A3 domain (residues 1811-1818) is shown in white, whereas the binding regions in the A2 domain (residues 558-565 and 698-710) are shown in dark and light blue, respectively (space-filling representations). These sites are in close vicinity, and are exposed at the same side of the molecule. The factor VIII A2 domain is required to induce significant changes within the factor IXa protease domain, indicating that it binds to the factor IXa protease domain. The A3 domain of factor VIII has been proposed to interact with the factor IXa light chain. Within the factor IXa light chain, residues 12, 64, 69, 78, 92, and 94 (see refs 150 to 155) are indicated (red, space-filling representation). These residues have been reported to be associated with an abnormal response to factor VIIIa in factor X activation.

Fig. 4.

Model of the factor VIII and factor IXa molecules. Shown are representiations of porcine factor IXa (Protein Data Bank accession code 1pfx) and the triplicated A-domains of human factor VIII (Hemophilia A web site, http://europium.mrc.rpms.ac.uk), which are derived from crystallography and homology modeling, respectively. Factor IXa binding region in the factor VIII A3 domain (residues 1811-1818) is shown in white, whereas the binding regions in the A2 domain (residues 558-565 and 698-710) are shown in dark and light blue, respectively (space-filling representations). These sites are in close vicinity, and are exposed at the same side of the molecule. The factor VIII A2 domain is required to induce significant changes within the factor IXa protease domain, indicating that it binds to the factor IXa protease domain. The A3 domain of factor VIII has been proposed to interact with the factor IXa light chain. Within the factor IXa light chain, residues 12, 64, 69, 78, 92, and 94 (see refs 150 to 155) are indicated (red, space-filling representation). These residues have been reported to be associated with an abnormal response to factor VIIIa in factor X activation.

Close modal

Assuming an interaction between the factor IXa protease domain and the factor VIII A2 domain, it seems conceivable that the A3 domain interacts with a region within the factor IXa light chain. This is in line with recent observations that the light chain of factor VIII binds to the light chain of factor IXa.138,149 Furthermore, mutations within the factor IXa light chain have been described that are associated with an abnormal response to factor VIIIa in factor X activation.150-154 These mutations are dispersed over the factor IXa light chain, indicating that multiple sites may contribute to binding of the factor VIII A3 domain. Alternatively, some mutations may destabilize the factor VIII binding site by affecting the conformation of the factor IXa light chain. This latter possibility has been reported for two distinct factor IX mutations.154 155 

Collectively, by combining biochemical data with the three-dimensional models of factor IXa and factor VIII, it appears that the factor VIII A2 domain binds to the factor IXa heavy chain, and the factor VIII A3 domain to the factor IXa light chain (Fig 4). It is of importance to realize that for factor VIII as well as for factor IXa the current models not fully represent the biologically active molecules. The factor IXa structure has been determined in the absence of calcium ions,143 which are obligatory for optimal exposure of the factor VIII light chain binding site and of the catalytic centre.133,155 The factor VIII model provided by Pemberton et al145 comprises the A domains only and lacks the B and C domains and the acidic domain spacers. It cannot be excluded that these domains affect the structure of the A domains. In addition, this factor VIII model does not distinguish between the inactive procofactor or activated factor VIII. It is obvious that both factor VIII species will have different structural properties, because only factor VIII, which is cleaved at specific positions, is able to stimulate factor IXa activity. Despite these restrictions, both the factor VIII and factor IXa model provide an important basis for proper selection of residues that may be investigated for their contribution in the assembly of the factor VIII–factor IX complex.

Factor IXa binding and hemophilia A.

Inspection of the hemophilia A database shows that several mutations have been reported that are in or close to the factor IXa binding sites: codons 558, 565, and 566, codons 698, 701, and 704 in the heavy chain, and codons 1789, 1796, 1823, and 1825 in the factor VIII light chain.49 It seems reasonable to assume that the bleeding tendency which is associated with these mutations is caused by suboptimal assembly of the factor IXa–factor VIIIa complex. With regard to the 558 and 566 mutation, this view has been supported by studies using recombinant factor VIII.156 Interestingly, a mutation outside the factor IXa–binding regions, ie, at codon Arg527, also has been reported to be associated with inefficient stimulation of factor IXa proteolytic activity.157,158 Examination of the three-dimensional model shows that Arg527 is located in the immediate vicinity of the factor IXa binding sequence 558 to 565.145 The exposure of this factor IXa binding site may be affected by substitution of Arg527. Alternatively, Arg527 may be part of an extensive factor IXa–binding interface, involving multiple sites of the A2 domain.

Mechanisms of inactivation.

Downregulation of the factor X–activating complex may involve inactivation or inhibition of either the enzyme factor IXa or the cofactor, factor VIIIa. Inactivation of the cofactor comprises two distinct pathways: proteolytic degradation and spontaneous dissociation. Once activated, factor VIII cofactor activity is rapidly lost.135,159-162 Compared with activated factor VIII, the procofactor is markedly more stabile, which is illustrated by its dissociation rate being 100-fold lower (kdiss ≈4 to 6 × 10−4 s−1 and 4 × 10−6 s−1 for factor VIIIa and factor VIII, respectively).47,163 The intrinsic instability of factor VIIIa can be attributed to the weak interaction between the A2 domain and the metal ion-linked A1/A3-C1-C2 dimer.164-166 The kd for this interaction is approximately 0.2 μmol/L.42 Because this value exceeds the factor VIII concentration in plasma 100- to 1,000-fold, equilibrium is in favor of the inactive, dissociated state of factor VIIIa.

Proteolytic degradation of factor VIIIa involves cleavages in the heavy chain at positions 336 and 562 by various enzymes, such as factor IXa, factor Xa, and APC.74-79,167-170 Cleavage at position 336 in factor VIIIa releases a1, the acidic sequence that interconnects the A1 and A2 domain. Because of this release, the A2 domain dissociates more rapidly from the factor VIIIa heterotrimer.115 This acidic spacer has been proposed to comprise a binding site for the substrate factor X,116,117indicating that release of this site results in impaired substrate binding. Thus, cleavage at Arg336 affects both intramolecular (A2 domain dissociation) and intermolecular (factor VIII–factor X) interactions. Arg562, which is part of the A2 domain sequence that comprises a factor IXa interactive site, is exclusively cleaved by APC.168 It seems conceivable that loss of cofactor activity due to cleavage at this site reflects the loss of the ability to interact with factor IXa.

One intriguing question is whether proteolytic degradation or spontaneous dissociation dominates the inactivation of factor VIII in vivo. At present the relative contribution of each mechanism to factor VIII inactivation is not fully understood, although some reports indicate that spontaneous dissociation is dominant.162,171,172 This view is underscored by the observation that dissociation of the factor VIIIa heterotrimer may be accelerated by binding of the A2 domain to the low-density lipoprotein receptor-related protein.173 To describe the process of factor VIIIa inactivation, it should further be considered that factor IXa plays a dual role. It stabilizes factor VIIIa by linking the A2 domain to the A3 domain,137,142,159 and protects factor VIII against inactivation by APC.136,167,174 On the other hand, under certain conditions factor IXa may inactivate factor VIIIa by cleavage at position 336,169 170 a site that is shared with factor Xa and APC. The fact that factor IXa is involved both in stabilization and in inactivation of factor VIII complicates a final assessment of the regulatory role of factor IXa in intrinsic factor Xa formation.

Defects in factor VIII inactivation.

Impaired inactivation of factor VIIIa or its homologue factor Va may be associated with a disturbed balance between procoagulant and anticoagulant systems. With respect to factor Va this view is supported by the notion that mutation at Arg506, a site that is cleaved by APC, predisposes to venous thromboembolism.175-178 It has been investigated whether patients displaying venous thromboembolism carry analogous mutations at the APC cleavage sites in factor VIII (ie, Arg336 and Arg562).179,180 However, this association has not been observed, which suggests that mutations at these positions are rare. Alternatively, such mutations may not predispose to thrombotic disorders, indicating that proteolytic inactivation of factor VIIIa is less important than inactivation of factor Va with regard to the hemostatic balance. This would be in agreement with the fact that murine factor VIII lacks the inactivation site at position 336.15 In addition, in vitro data using genetically engineered factor VIII with mutations in the APC cleavage sites showed that these mutations were not associated with reduced clotting times in APC-resistance assays.181 

Although APC-resistant factor VIII molecules have not been identified in patients, the possibility remains open that APC-resistance may modulate factor VIII inactivation in an indirect manner. Inactivation of factor VIII by APC is enhanced in the presence of the APC-cofactor protein S.74,167 Several investigators have reported that the factor V procofactor enhances the cofactor effect of protein S in factor VIII inactivation.162,182,183 This link between factor V and factor VIII inactivation becomes even more apparent by the finding of Váradi et al,183 who reported that factor V, which carries the Arg506 to Gln mutation, has impaired cofactor activity in APC– and protein S–dependent factor VIII inactivation. The physiological significance of this observation remains unclear. However, the possibility that APC-resistance also affects factor VIII inactivation is challenging and deserves further study.

Another intriguing finding is that APC-resistant factor V has the potential to bypass the absence of factor VIII activity to some extent in in vitro thrombin generation studies.184 Therefore, it cannot be excluded that hemophilic patients which carry the factor V Arg506 to Gln mutation display a less severe phenotype than expected. This indeed has been shown for some hemophilic patients as described by Nichols et al.185 However, Arbini et al186 did not find an association between the presence of APC-resistant factor V and the severity of the bleeding tendency in a population of 295 hemophilic patients. The ability of APC-resistant factor V to bypass factor VIII deficiency may be restricted to specific, thus far unrecognized conditions.

At present, little is known about the mechanism by which factor VIII is cleared from the circulation. Obviously, vWF serves an important role, because in patients with severe vWD the factor VIII half-life is considerably decreased.83,86-88 As vWF protects factor VIII against proteolytic degradation in vitro (see Assembly of the Factor VIII–vWF Complex section), it cannot be excluded that the decreased half-life in the absence of vWF factor is associated with proteolytic degradation of factor VIII. However, experimental data in support of this possibility are lacking thus far. Another explanation for the rapid clearance could be binding of noncomplexed factor VIII to the surface of cells. In this respect it is of interest to mention that hepatic endothelial cells have been reported to contain factor VIII protein, but not its mRNA (see Biosynthesis and Secretion of Factor VIII section). It may be noteworthy that the copper-binding protein ceruloplasmin, which is structurally related to the factor VIII A domains, is internalized in the hepatic endothelial cells through a receptor-mediated process.187 It seems possible that factor VIII is taken up by hepatic endothelium by a similar mechanism.

Because the effect of factor V proteolysis on its survival has been investigated in a nonhuman primate model,188 it is of interest to compare factor VIII survival with that of factor V. Whereas the half-life of the factor V procofactor is approximately 14 hours, the half-life of its thrombin-activated derivative is dramatically different. The heavy and light chain are cleared very rapidly (t1/2 < 20 minutes). The half-life for factor V light chain is remarkably close to the half-life of 10 minutes reported for the isolated light chain of factor VIII after infusion into hemophilic dogs.189 Perhaps, clearance of both proteins involves a similar mechanism. Clearance of the factor V activation peptide, ie, the B domain, is considerably slower (t1/2 > 30 hours),188 suggesting a previously unrecognized role of this domain in preventing premature clearance of factor V. Whether this is also true for the factor VIII B domain is not clear. The half-lifes of normal factor VIII and B-domain deleted factor VIII are similar in hemophilic patients190 as well in hemophilic dogs,87,191 192 indicating that the B domain does not contribute to factor VIII survival. However, it is important to note that these data reflect factor VIII survival in the presence of vWF. Therefore, it would be of interest to investigate survival of B-domainless factor VIII in patients with vWD type 3.

Structure-function studies have contributed significantly to our current understanding of factor VIII biology and the molecular background of hemophilia A. As such this has provided the basis for the development of second generation recombinant factor VIII molecules that may find a therapeutical application. Pertinent to this point are the B-domain–deleted factor VIII variants, which are subject to clinical (factor VIII-SQ)190 or preclinical108,191,192testing. Furthermore, various factor VIII variants have been designed which in the future may be particular useful in the treatment of hemophilia A. Variants have been described which are less prone to inhibitor neutralization.193,194 Other examples include variants with enhanced hemostatic potency114 or stability.195 

Despite the rapid accumulation of information regarding the structure and function of factor VIII, a number of questions remain to be answered. For instance, it is still unclear why factor VIII becomes a potent cofactor of factor IXa once it is cleaved within the heavy and light chain. What is the structural basis for such a dramatic increase in biological activity? How does factor VIIIa, together with the membrane surface, push factor IXa into an extremely potent configuration? It seems obvious that studies on three-dimensional modeling techniques based on crystal structures of factor VIII or factor VIII fragments complexed with their ligands (eg, factor IX) should provide a solid basis for a better understanding of the molecular aspects of factor VIII function and dysfunction. Because both factor VIII and factor IX are now potentially available in substantial quantities, determination of the crystal structure of complexes comprising wild-type or mutant proteins should be feasible and undoubtedly will lead to significant advances in this field. This approach will also facilitate the design of antagonistic inhibitors of the factor VIII system, which could provide novel anticoagulants for the treatment of thrombotic disorders.

The question of why exposure to factor VIII concentrates is associated with allo-immune reponses in some of the hemophilia A patients remains unanswered. What is the mechanism that causes these adverse reactions? Why and how is the immune system challenged under these conditions? Why do certain specific polypeptide regions of the factor VIII molecule play a prominent role in these immune reactions? Finally, little is known about the origin and expression of factor VIII at the cellular level. Which mechanisms trigger the specific cellular responses that govern elevations of the factor VIII plasma level under a variety of clinical conditions? Similarly, the mechanism of factor VIII clearance is an unexplored subject. Finding the answers to these questions is not only of fundamental, merely scientific appeal, but also has the potential of further improving our current strategies for the treatment of hemophilia A.

We express our gratitude to Drs W.G. van Aken, J. Voorberg, O.D. Christophe, and K. Fijnvandraat for helpful discussions and critical reading of the manuscript. We also thank Dr G. Kemball-Cook for providing the coordinates of the factor VIII.

1
Rosner
F
Hemophilia in the talmud and rabbinic writings.
Ann Intern Med
70
1969
833
2
Sadler
JE
Davie
EW
Hemophilia A, Hemophilia B, and von Willebrand’s disease
The Molecular Basis of Blood Diseases.
Stamatoyannopoulos
G
Nienhuis
A
Leder
P
Majerus
P
1987
576
Saunders
Philadelphia, PA
3
Patek
AJ
Taylor
FHL
Hemophilia. II. Some properties of a substance obtained from normal plasma effective in accelerating the clotting of hemophilic blood.
J Clin Invest
16
1937
113
4
Mann
KG
Nesheim
ME
Church
WR
Haley
P
Krishnaswamy
S
Surface dependent reactions of the vitamin K-dependent enzyme complexes.
Blood
76
1990
1
5
Gitschier
J
Wood
WI
Goralka
TM
Wion
KL
Chen
EY
Eaton
DH
Vehar
GA
Capon
DJ
Lawn
RM
Characterization of the human factor VIII gene.
Nature
312
1984
326
6
Wood
WI
Capon
DJ
Simonson
CC
Eaton
DL
Gitschier
J
Keyt
B
Seeburg
PH
Smith
DH
Hollingshead
P
Wion
KL
Delwart
E
Tuddenham
EGD
Vehar
GA
Lawn
RM
Expression of active human factor VIII from recombinant DNA clones.
Nature
312
1984
330
7
Vehar
GA
Keyt
B
Eaton
D
Rodriguez
H
O’Brien
DP
Rotblat
F
Oppermann
H
Keck
R
Wood
WI
Harkins
RN
Tuddenham
EGD
Lawn
RM
Capon
DJ
Structure of human factor VIII.
Nature
312
1984
337
8
Toole
JJ
Knopf
JL
Wozney
JM
Sultzman
LA
Bueker
JL
Pittman
DD
Kaufman
RD
Brown
E
Shoemaker
C
Orr
EC
Amphlett
GW
Foster
WB
Coe
ML
Knutson
GJ
Fass
DN
Hewick
RM
Molecular cloning of a cDNA encoding human antihaemophilic factor.
Nature
312
1984
342
9
Church
WR
Jernigan
RL
Toole
J
Hewick
RM
Knopf
J
Knutson
GJ
Nesheim
ME
Mann
KG
Fass
DN
Coagulation factors V and VIII and ceruloplasmin constitute a family of structurally related proteins.
Proc Natl Acad Sci USA
81
1984
6934
10
Stubbs
JD
Lekutis
C
Singer
KL
Bui
A
Yuzuki
D
Srinivasan
U
Parry
G
cDNA cloning of a mouse mammary epithelial cell surface protein reveals the existence of epidermal growth factor-like domains linked to factor VIII-like sequences.
Proc Natl Acad Sci USA
87
1990
8417
11
Larocca
D
Peterson
JA
Urrea
R
Kuniyoshi
J
Bistrain
AM
Ceriani
RL
A Mr 46,000 human milk fat globule protein that is highly expressed in human breast tumors contains factor VIII-like domains.
Cancer Res
51
1991
4994
12
Takagi
S
Hirata
T
Agata
K
Mochii
M
Eguchi
G
Fujisawa
H
The A5 antigen, a candidate for the neuron recognition molecule, has homologies to complement proteins and coagulation factors.
Neuron
7
1991
295
13
Wion
KL
Kelly
D
Summerfield
JA
Tuddenham
EGD
Lawn
RM
Distribution of factor VIII mRNA and antigen in human liver and other tissues.
Nature
317
1985
726
14
Levinson
B
Kenwrick
S
Gamel
P
Fisher
K
Gitschier
J
Evidence for a third transcript from the human factor VIII gene.
Genomics
14
1992
585
15
Elder
B
Lakich
D
Gitschier
J
Sequence of the murine factor VIII cDNA.
Genomics
16
1993
374
16
Groth
CG
Hathaway
WE
Gustafsson
A
Geis
WP
Putnam
CW
Bjorken
C
Porter
KA
Starzl
TE
Correction of coagulation in the hemophilic dog by transplantaion of lymphatic tissue.
Surgery
75
1974
725
17
Veltkamp
JJ
Asfaou
E
van der Torren
E
van der Does
JA
van Tilburg
NH
Pauwels
EKJ
Extrahepatic factor VIII synthesis. Lung transplants in hemophilic dogs.
Transplantation
18
1974
56
18
Shaw
E
Giddings
JC
Peake
IR
Bloom
AL
Synthesis of procoagulant factor VIII, factor VIII-related antigen, and other coagulation factors by the isolated perfused rat liver.
Br J Haematol
41
1979
585
19
Owen
CA
Bowie
EJW
Fass
DN
Generation of factor VIII coagulant activity by isolated, perfused neonatal pig livers and adult rat livers.
Br J Haematol
43
1979
307
20
Marchioro
TL
Hougie
C
Ragde
H
Epstein
RB
Thomas
ED
Hemophilia: Role of organ homografts.
Science
163
1969
188
21
Lewis
JH
Bontempo
FA
Spero
JA
Gorenc
TJ
Ragni
MV
Starzl
TE
Liver transplantation in a hemophiliac.
N Engl J Med
312
1985
1189
22
Bontempo
FA
Lewis
JH
Gorenc
TJ
Spero
JA
Ragni
MV
Scott
JP
Starzl
TE
Liver transplantation in hemophilia A.
Blood
69
1987
1721
23
Figueiredo
MS
Brownlee
GG
cis-Acting elements and transcription factors involved in the promotor activity of the human factor VIII gene.
J Biol Chem
270
1995
11828
24
Zelechowska
MG
van Mourik
JA
Brodniewicz-Proba
T
Ultrastructural localization of factor VIII procoagulant antigen in liver hepatocytes.
Nature
317
1985
729
25
Stel
HV
van der Kwast
TH
Veerman
ECI
Detection of factor VIII/coagulant antigen in human liver tissue.
Nature
303
1983
530
26
van der Kwast
TH
Stel
HV
Cristen
E
Bertina
RM
Veerman
ECI
Localization of factor VIII-procoagulant antigen: An immunohistological survey of the human body using monoclonal antibodies.
Blood
67
1986
222
27
Kadhom
N
Wolfrom
C
Gautier
M
Allain
JP
Frommel
D
Factor VIII procoagulant antigen in human tissues.
Thromb Haemost
59
1988
289
28
Kaufman
RJ
Pipe
SW
Tagliavacca
L
Swaroop
M
Moussalli
M
Biosynthesis, assembly and secretion of coagulation factor VIII.
Blood Coagul Fibrinol
8
1997
S3
(suppl 2)
29
Kaufman
RJ
Wasley
LC
Davies
MV
Wise
RJ
Israel
DI
Dorner
AJ
Effect of von Willebrand factor coexpression on the synthesis and secretion of factor VIII in Chinese hamster ovary cells.
Mol Cell Biol
9
1989
1233
30
Dorner
AJ
Bole
DG
Kaufman
RJ
The relationship of N-linked glycosylation and heavy chain-binding protein association with the secretion of glycoproteins.
J Cell Biol
105
1987
2665
31
Dorner
AJ
Wasley
LC
Kaufman
RJ
Increased synthesis of secreted proteins induces expression of glucose regulated proteins in butyrate treated CHO cells.
J Biol Chem
264
1989
20602
32
Marquette
KA
Pittman
DD
Kaufman
RJ
A 110 amino acid region within the A1 domain of coagulation factor VIII inhibits secretion from mammalian cells.
J Biol Chem
270
1995
10297
33
Swaroop
M
Moussalli
M
Pipe
SW
Kaufman
RJ
Mutagenesis of a potential immunoglobulin-binding-protein-binding site enhances secretion of coagulation factor VIII.
J Biol Chem
272
1997
24121
34
Pipe
SW
Morris
JA
Shah
J
Kaufman
RJ
Differential interaction of coagulation factor VIII and factor V with protein chaperones calnexin and calreticulin.
J Biol Chem
273
1998
8537
35
Nichols
WC
Seligsohn
U
Zivelin
A
Terry
VH
Hertel
CE
Wheatly
MA
Moussalli
MJ
Hauri
HP
Ciavarella
N
Kaufman
RJ
Ginsburg
D
Mutations in the ER-Golgi intermediate compartment protein ERGIC-53 cause combined deficiency of coagulation factors V and VIII.
Cell
93
1998
61
36
Kaufman
RJ
Wasley
LC
Dorner
AJ
Synthesis, processing, and secretion of recombinant human factor VIII expressed in mammalian cells.
J Biol Chem
263
1988
6352
37
van de Ven
WJM
Voorberg
J
Fontijn
R
Pannekoek
H
van den Ouweland
AMW
van Duijnhoven
HLP
Roebroek
AJM
Siezen
RJ
Furin is a subtilisin-like proprotein processing enzyme in higher eukaryotes.
Mol Biol Rep
14
1990
265
38
Hutton
JC
Subtilisin-like proteinases involved in the activation of proproteins of the eukaryotic secretory pathway.
Curr Opin Cell Biol
2
1990
1131
39
Barr
PJ
Mammalian subtilisins: The long-sought dibasic processing endoproteases.
Cell
66
1991
1
40
Fass
DN
Knutson
GJ
Katzmann
JA
Monoclonal antibodies to porcine factor VIII procoagulant and their use in the isolation of active coagulant protein.
Blood
59
1982
594
41
Fay
PJ
Anderson
MT
Chavin
SI
Marder
VJ
The heterodimeric structure of human factor VIII and changes produced by thrombin interaction.
Biochim Biophys Acta
871
1986
268
42
Fay
PJ
Smudzin
TM
Characterization of the interaction between the A2 subunit and A1/A3-C1-C2 dimer in human factor VIIIa.
J Biol Chem
267
1992
13246
43
Bihoreau
N
Pin
S
de Kersabiec
AM
Vidot
F
Fontaine-Aupart
MP
Copper-ion identification in the active and inactive forms of plasma-derived FVIII and recombinant FVIII-▵II.
Eur J Biochem
220
1994
41
44
Tagliavacca
L
Moon
N
Dunham
WR
Kaufman
RJ
Identification and functional requirement of Cu(I) and its ligands within coagulation factor VIII.
J Biol Chem
272
1997
27428
45
Fay
PJ
Reconstitution of human factor VIII from isolated subunits.
Arch Biochem Biophys
262
1988
525
46
Nordfang
O
Ezban
M
Generation of active coagulation factor VIII from isolated subunits.
J Biol Chem
263
1988
1115
47
Donath
MJSH
Lenting
PJ
van Mourik
JA
Mertens
K
The role of cleavages at positions Arg1689 and Arg1721 in subunit interaction and activation of human factor VIII.
J Biol Chem
270
1995
3648
47a
Sudkahar
K
Fay
PJ
Effects of copper on the structure and function of factor VIII subunits: Evidence for an auxiliary role for copper ions in cofactor activity.
Biochemistry
37
1998
6874
48
Gitschier
J
Wood
WI
Shuman
MA
Lawn
RM
Identification of a missense mutation in the factor VIII gene of a mild hemophiliac.
Science
232
1986
1415
49
Kemball-Cook G, Tuddenham EGD: The factor VIII mutation database on the world wide web: the haemophilia A mutation, search, test and resource site. Hamsters update (version 3.0,http://europium.mrc.rpms.ac.uk). Nucleic Acids Res 25:128, 1997
50
Voorberg
J
de Laaf
RTM
Koster
PM
van Mourik
JA
Intracellular retention of a factor VIII protein with an Arg2307→Gln mutation as a cause of haemophilia A.
Biochem J
318
1996
931
51
Pipe
SW
Kaufman
RJ
Factor VIII C2 domain missense mutations exhibit defective trafficking of biological functional proteins.
J Biol Chem
271
1996
25671
52
Nichols
WC
Seligsohn
U
Zivelin
A
Terry
V
Arnold
ND
Siemieniak
DR
Kaufman
RJ
Ginsburg
D
Linkage of combined factors V and VIII deficiency to chromosome 18q by homozygosity mapping.
J Clin Invest
99
1997
596
53
Neerman-Arbez
M
Antonarakis
SE
Blouin
JL
Zeinali
S
Akhtari
M
Afshar
Y
Tuddenham
EG
The locus for combined factor V-factor VIII deficiency (F5F8D) maps to 18q21, between D18S849 and D18S1103.
Am J Hum Genet
61
1997
143
54
Lollar
P
Hill-Eubanks
DC
Parker
CG
Association of the factor VIII light chain with von Willebrand factor.
J Biol Chem
263
1988
10451
55
Vlot
AJ
Koppelman
SJ
van den Berg
MH
Bouma
BN
Sixma
JJ
The affinity and stoichiometry of binding of human factor VIII to von Willebrand factor.
Blood
85
1995
3150
56
Leyte
A
Verbeet
MP
Brodniewicz-Proba
T
van Mourik
JA
Mertens
K
The interaction between human blood coagulation factor VIII and von Willebrand factor.
Biochem J
257
1989
679
57
Saenko
EL
Scandella
D
The acidic region of the factor VIII light chain and the C2 domain together form the high affinity binding site for von Willebrand factor.
J Biol Chem
272
1997
18007
58
Foster
PA
Fulcher
CA
Houghten
RA
Zimmerman
TS
An immunogenic region within residues Val1670-Glu1684 of the factor VIII light chain induces antibodies which inhibit binding of factor VIII to von Willebrand factor.
J Biol Chem
263
1988
5230
59
Precub
JW
Kline
BC
Fass
DN
A monoclonal antibody to factor VIII inhibits von Willebrand factor binding and thrombin cleavage.
Blood
77
1991
1929
60
Shima
M
Scandella
D
Yoshioka
A
Nakai
H
Tanaka
I
Kamisau
S
Terada
S
Fukui
H
A factor VIII neutralizing monoclonal antibody and a human inhibitor alloantibody recognizing epitopes in the C2 domain inhibit factor VIII binding to von Willebrand factor and to phosphatidylserine.
Thromb Haemost
69
1993
240
61
Shima
M
Nakai
H
Scandella
D
Tanaka
I
Sawamoto
Y
Kamisue
S
Morichika
S
Murakami
T
Yoshioka
A
Common inhibitory effects of human anti-C2 domain inhibitor alloantibodies on factor VIII binding to von Willebrand factor.
Br J Haematol
91
1995
714
62
Saenko
EV
Shima
M
Rajalakshmi
KJ
Scandella
D
A role for the C2 domain of factor VIII in binding to von Willebrand factor.
J Biol Chem
269
1994
11601
63
Leyte
A
van Schijndel
HB
Niehrs
C
Huttner
WB
Verbeet
MP
Mertens
K
van Mourik
JA
Sulphation of Tyr1680 of human blood coagulation factor VIII is essential for the interaction of factor VIII with von Willebrand factor.
J Biol Chem
266
1991
740
64
Meulien
P
Faure
T
Mischler
F
Harrer
H
Ulrich
P
Bouderbala
B
Dott
K
Marie
MS
Mazurier
C
Wiesel
ML
van der Pol
H
Cazenave
JP
Courtney
M
Pavirani
A
A new recombinant procoagulant protein derived from the cDNA encoding human factor VIII.
Protein Eng
2
1988
301
65
Pittman
DD
Wang
JH
Kaufman
RJ
Identification and functional importance of tyrosine sulfate residues within recombinant factor VIII.
Biochemistry
31
1992
3315
66
Michnick
DA
Pittman
DD
Wise
RJ
Kaufman
RJ
Identification of individual tyrosine sulfation sites within factor VIII required for optimal activity and efficient thrombin cleavage.
J Biol Chem
269
1994
20095
67
Lenting
PJ
Donath
MJSH
van Mourik
JA
Mertens
K
Identification of a binding site for blood coagulation factor IXa on the light chain of human factor VIII.
J Biol Chem
269
1994
7150
68
Foster
PA
Fulcher
CA
Houghten
RA
Zimmerman
TS
Synthetic factor VIII peptides with amino acid sequences contained within the C2 domain of factor VIII inhibit factor VIII binding to phosphatidyl serine.
Blood
75
1990
1999
69
Saenko
EL
Shima
M
Gilbert
GE
Scandella
D
Slowed release of thrombin-cleaved factor VIII from von Willebrand factor by a monoclonal and a human antibody is a novel mechanism for factor VIII inhibition.
J Biol Chem
271
1996
27424
70
Andersson
LO
Brown
JE
Interaction of factor VIII-von Willebrand factor with phospholipid vescicles.
Biochem J
200
1981
161
71
Nesheim
M
Pittman
DD
Giles
AR
Fass
DN
Wang
JH
Slonosky
D
Kaufman
RJ
The effect of plasma von Willebrand factor on the binding of human factor VIII to thrombin-activated platelets.
J Biol Chem
266
1991
17815
72
Gilbert
GE
Drinkwater
D
Barter
S
Clouse
SB
Specificity of phosphatidylserine-containing membrane binding sites for factor VIII.
J Biol Chem
267
1992
15861
73
Saenko
EL
Scandella
D
A mechanism for inhibition of factor VIII binding to phospholipid by von Willebrand factor.
J Biol Chem
270
1995
13826
74
Koedam
J
Meijers
J
Sixma
J
Bouma
B
Inactivation of human factor VIII by activated protein C: Cofactor activity of protein S and protective effect of von Willebrand factor.
J Clin Invest
82
1988
1236
75
Rick
ME
Esmon
N
Krizek
D
Factor IXa and von Willebrand factor modify the inactivation of factor VIII by activated protein C.
J Lab Clin Med
115
1990
415
76
Koedam
JA
Hamer
RJ
Beeser-Visser
NH
Bouma
BN
Sixma
JJ
The effect of von Willebrand factor on activation of factor VIII by factor Xa.
Eur J Biochem
189
1990
229
77
Fay
PJ
Coumans
J-V
Walker
FJ
von Willebrand factor mediates protection of factor VIII from activated protein C-catalysed inactivation.
J Biol Chem
266
1991
2172
78
Fay
PJ
Walker
FJ
Inactivation of human factor VIII by activated protein C: Evidence hat the factor VIII light chain contains the activated protein C binding site.
Biochim Biophys Acta
994
1989
142
79
Eaton
D
Rodriguez
H
Vehar
GA
Proteolytic processing of human factor VIII.
Biochemistry
25
1986
505
80
Hill-Eubanks
DC
Parker
CG
Lollar
P
Differential proteolytic activation of factor VIII-von Willebrand factor complex by thrombin.
Proc Natl Acad Sci USA
86
1989
6508
81
Esmon
CT
Lollar
P
Involvement of thrombin anion-binding exosites 1 and 2 in the activation of factor V and factor VIII.
J Biol Chem
271
1996
13882
82
Weiss
HJ
Sussman
II
Hoyer
LW
Stabilization of factor VIII in plasma by the von Willebrand factor.
J Clin Invest
60
1977
390
83
Tuddenham
EGD
Lane
RS
Rotblat
F
Johnson
AJ
Snape
TJ
Middleton
S
Kernoff
PBA
Response to infusions of polyelectrolyte fractionated human factor VIII concentrate in human haemophilia A and von Willebrand’s disease.
Br J Haematol
52
1982
259
84
Wise
RJ
Dorner
AJ
Krane
M
Pittman
DD
Kaufman
RJ
The role of von Willebrand factor multimers and propeptide cleavage in binding and stabilization of factor VIII.
J Biol Chem
266
1991
21948
85
Mannucci
PM
Tenconi
PM
Castaman
G
Rodeghiero
F
Comparison of four virus-inactivated plasma concentrates for treatment of severe von Willebrand disease: A cross-over randomized trial.
Blood
79
1992
3130
86
Over
J
Sixma
JJ
Bouma
BN
Vlooswijk
RAA
Beeser-Visser
NH
Survival of factor VIII in von Willebrand’s disease.
J Lab Clin Med
97
1981
332
87
Brinkhous
KM
Sandberg
H
Garris
JB
Mattson
C
Palm
M
Griggs
T
Read
MS
Purified human factor VIII procoagulant protein: Comparative hemostatic response after infusions into hemophilic and von Willebrand disease dogs.
Proc Natl Acad Sci USA
82
1985
8752
88
Morfini
M
Mannucci
PM
Tenconi
PM
Longo
G
Mazzucconi
MG
Rodeghiero
F
Ciavarella
N
De Rosa
V
Arter
A
Pharmacokinetics of monoclonally-purified and recombinant factor VIII in patients with severe von Willebrand disease.
Thromb Haemost
70
1993
270
89
Nishino
M
Girma
JP
Rothschild
C
Fressinaud
E
Meyer
D
New variant of von Willebrand disease with defective binding to factor VIII.
Blood
74
1989
1591
90
Mazurier
C
Diveval
J
Jorieux
S
Delobel
J
Goudemand
M
A new von Willebrand factor defect in a patient with factor VIII deficiency but with normal levels and multimeric patterns of both plasma and platelet von Willebrand factor.
Blood
75
1990
20
91
Higuchi
M
Wong
C
Kochhan
L
Aronis
S
Kasper
CK
Kazazian
HH
Antonarakis
SE
Mutations in the factor VIII gene by direct sequencing of amplified genomic DNA.
Genomics
6
1990
65
92
Traystmann
MD
Higuchi
M
Kasper
CK
Antonarakis
SE
Kazazian
HH
Use of denaturating gradient gel electrophoresis to detect point mutations in the factor VIII gene.
Genomics
6
1990
293
93
Vehar
G
Davie
EW
Preparation and properties of bovine FVIII (anti-hemophilic factor).
Biochemistry
19
1980
401
94
Fass
DN
Knutson
GJ
Katzmann
J
Monoclonal antibodies to porcine factor VIII coagulant and their use in the isolation of active coagulant protein.
Blood
59
1982
594
95
Fulcher
C
Zimmerman
T
Characterization of the human FVIII procoagulant protein with a heterologous precipitating antibody.
Proc Natl Acad Sci USA
79
1982
1648
96
Rotblat
F
O’Brien
SDP
O’Brien
FJ
Goodall
AH
Tuddenham
EGD
Purification of factor VIII:C and its characterization by western blotting using monoclonal antibodies.
Biochemistry
24
1985
4294
97
Pittman
DD
Kaufman
RJ
Proteolytic requirements for thrombin activation of anti-haemophilic factor (FVIII).
Proc Natl Acad Sci USA
85
1988
2429
98
Lollar
P
Knutson
GJ
Fass
DN
Activation of porcine FVIII:C by thrombin and factor Xa.
Biochemistry
24
1985
8056
99
Hoyer
LW
Trabold
NC
The effect of thrombin on human factor VIII.
J Lab Clin Med
97
1981
50
100
Mertens
K
Bertina
RM
Activation of human blood coagulation factor VIII by activated factor X, the common product of the intrinsic and the extrinsic pathway of blood coagulation.
Thromb Haemost
47
1982
96
101
Griffith
MJ
Reisner
H
Lundblad
R
Roberts
HR
Measurement of human factor IXa activity in an isolated factor X activating system.
Thromb Res
27
1982
289
102
Neuenschwander
PF
Jesty
J
Thrombin-activated and factor Xa-activated factor VIII: Differences in cofactor activity and decay rate.
Arch Biochem Biophys
296
1992
426
103
Parker
ET
Pohl
J
Blackburn
MN
Lollar
P
Subunit structure and function of porcine factor Xa-activated factor VIII.
Biochemistry
36
1997
9365
104
Regan
LM
Fay
PJ
Cleavage of factor VIII light chain is required for maximal generation of factor VIIIa activity.
J Biol Chem
270
1995
8546
105
Hortin
G
Folz
R
Gordon
JI
Strauss
AW
Characterization of sites of tyrosine sulfation in proteins and criteria for predicting their occurence.
Biochem Biophys Res Commun
141
1986
326
106
Huttner
WB
Tyrosine sulfation and the secretory pathway.
Annu Rev Physiol
50
1988
363
107
Hortin
GL
Benutto
BM
Inhibition of thrombin clotting activity by synthetic peptide segments of its inhibitors and substrates.
Biochem Biophys Res Commun
169
1990
437
108
Donath
MJSH
de Laaf
RTM
Biessels
PTM
Lenting
PJ
van de Loo
JW
van Mourik
JA
Voorberg
J
Mertens
K
Characterization of des-(741-1668)-factor VIII, a single-chain factor VIII variant with a fusion site susceptible to proteolysis by thrombin and factor Xa.
Biochem J
312
1995
49
109
Bihoreau
N
Paolantonacci
P
Bardelle
C
Fontaine-Aupart
MP
Krishnan
S
Yon
J
Romet-Lemonne
JL
Structural and functional characterization of factor VIII-▵II, a new recombinant factor VIII lacking most of the B-domain.
Biochem J
277
1991
23
110
Kjalke
M
Heding
A
Talbo
G
Persson
E
Thomsen
J
Ezban
M
Amino acid residues 721-729 are required for full factor VIII activity.
Eur J Biochem
234
1995
773
111
Mikkelsen
J
Thomsen
J
Ezban
M
Heterogeneity in tyrosine sulfation of chinese hamster ovary cell produced recombinant factor VIII.
Biochemistry
30
1991
1533
112
Donath
MJSH
Lenting
PJ
van Mourik
JA
Mertens
K
Kinetics of factor VIII light-chain cleavage by thrombin and factor Xa.
Eur J Biochem
240
1996
365
113
Leyte
A
Mertens
K
Distel
B
Evers
RF
De Keyzer-Nellen
MJM
Groenen-van Dooren
MMCL
De Bruin
J
Pannekoek
H
van Mourik
JA
Verbeet
MP
Inhibition of human blood coagulation factor VIII by monoclonal antibodies.
Biochem J
263
1989
187
114
Voorberg
J
van Stempvoort
G
Klaasse Bos
JM
Mertens
K
van Mourik
JA
Donath
MJSH
Enhanced thrombin sensitivity of a factor VIII-heparin cofactor II hybrid.
J Biol Chem
271
1996
20985
115
Fay
PJ
Haidaris
PJ
Huggins
CF
Role of the COOH-terminal acidic region of A1 subunit in A2 subunit retention in human factor VIIIa.
J Biol Chem
268
1993
17861
116
Regan
LM
O’Brien
LM
Beattie
TL
Sudhakar
K
Walker
FJ
Fay
PJ
Activated protein C-catalyzed proteolysis of factor VIIIa alters its interactions with factor Xase.
J Biol Chem
271
1996
3982
117
Lapan
KA
Fay
PJ
Localization of a factor X interactive site in the A1 subunit of factor VIIIa.
J Biol Chem
272
1997
2082
118
Gitschier
J
Kogan
S
Levinson
B
Tuddenham
EGD
Mutations of factor VIII cleavage sites in hemophilia A.
Blood
72
1988
1022
119
O’Brien
DP
Tuddenham
EGD
Purification and characterization of factor VIII 1,689-Cys: A nonfunctional cofactor occurring in a patient with severe hemophilia A.
Blood
73
1989
2117
120
Shima
M
Ware
J
Yoshioka
A
Fukui
H
Fulcher
CA
An arginine to cysteine amino acid substitution at a critical thrombin cleavage site in a dysfunctional factor VIII molecule.
Blood
74
1989
1612
121
Arai
M
Higuchi
M
Antonarakis
SE
Kazazian
HH
Philips
JA
Janco
RL
Hoyer
LW
Characterization of a thrombin cleavage site mutation (Arg 1689 to Cys) in the factor VIII gene of two unrelated patients with cross-reacting material-positive hemophilia A.
Blood
75
1990
384
122
Pattinson
JK
McVey
JH
Boon
M
Ajani
A
Tuddenham
EGD
CRM+ haemophilia A due to a missense mutation (372→Cys) at the internal heavy chain thrombin cleavage site.
Br J Haematol
75
1990
73
123
Acquila
M
Caprino
D
Pecorara
M
Baudo
F
Morfini
M
Mori
PG
Two novel mutations at 373 codon of FVIII gene detected by DGGE.
Thromb Haemost
69
1993
392
124
Johnson
DJ
Pemberton
S
Acquila
M
Mori
PG
Tuddenham
EG
O’Brien
DP
FVIII S373L: Mutation at P1’ site confers thrombin cleavage resistance, causing mild haemophilia A.
Thromb Haemost
71
1994
428
125
Aly
AM
Arai
M
Hoyer
LW
Cysteamine enhances the procoagulant activity of factor VIII-East Hartford, a dysfunctional protein due to a light chain thrombin cleavage site mutation (arginine-1689 to cysteine).
J Clin Invest
89
1992
1375
126
Aly
AM
Hoyer
LW
Factor VIII-East Hartford (arginine 1689 to cysteine) has procoagulant activity when separated from von Willebrand factor.
J Clin Invest
89
1992
1382
127
Duffy
EJ
Parker
ET
Mutucumarana
VP
Johnson
AE
Lollar
P
Binding of factor VIIIa and factor VIII to factor IXa on phospholipid vesicles.
J Biol Chem
267
1992
17006
128
Bloom
JW
The interaction of rDNA factor VIII, factor VIII-des-(797-1562), and factor VIII-des-(797-1562)-derived peptides with phospholipid.
Thromb Res
48
1987
439
129
Gilbert
GE
Furie
BC
Furie
B
Binding of human factor VIII to phospholipids.
J Biol Chem
265
1990
815
130
Gilbert
GE
Drinkwater
D
Specific membrane binding of factor VIII is mediated by O-phospho-L-serine, a moiety of phosphatidylserine.
Biochemistry
32
1993
9577
131
Spaargaren
J
Giessen
PLA
Janssen
MP
Voorberg
J
Willems
GM
van Mourik
JA
Binding of blood coagulation factor VIII and its light chain to phosphatidylserine/phosphatidylcholine bilayers as measured by ellipsometry.
Biochem J
310
1995
539
132
Gilbert
GE
Arena
AA
Activation of the factor VIIIa-factor IXa enzyme complex of blood coagulation by membranes containing phosphatidyl-L-serine.
J Biol Chem
271
1996
11120
133
Mathur
A
Zhong
D
Sabharwal
AK
Smith
KJ
Bajaj
SP
Interaction of factor IXa with factor VIIIa.
J Biol Chem
272
1997
23418
134
Lamphear
BJ
Fay
PJ
Factor IXa enhances reconstitution of factor VIIIa from isolated A2 subunit and A1/A3-C1-C2 dimer.
J Biol Chem
267
1992
3725
135
Curtis
JE
Helgerson
SL
Parker
ET
Lollar
P
Isolation and characterization of thrombin-activated human factor VIII.
J Biol Chem
269
1994
6246
136
Regan
LM
Lamphear
BJ
Huggins
CF
Walker
FJ
Fay
PJ
Factor IXa protects factor VIIIa from activated protein C.
J Biol Chem
269
1994
9445
137
Fay
PJ
Beattie
T
Huggins
CF
Regan
LM
Factor VIIIa A2 subunit residues 558-565 represent a factor IXa interactive site.
J Biol Chem
269
1994
20522
138
O’Brien
LM
Medved
LV
Fay
PJ
Localization of factor IXa and factor VIIIa interactive sites.
J Biol Chem
270
1995
27087
139
Mutucumarana
VP
Duffy
EJ
Lollar
P
Johnson
AE
The active site of factor IXa is located far above the membrane surface and its conformation is altered upon association with factor VIIIa.
J Biol Chem
267
1992
17012
140
Jorquera
JI
McClintock
RA
Roberts
JR
MacDonald
MJ
Houghten
RA
Fulcher
CA
Synthetic peptides derived from residues 698 to 710 of factor VIII inhibit factor IXa activity.
Circulation
86
1992
685
(abstr)
141
Liles
DK
Monroe
DM
Roberts
HR
The factor VIII peptide consisting of amino acids 698 to 712 enhances factor IXa cleavage of factor X.
Blood
90
1997
463a
(abstr, suppl 1)
142
Lenting
PJ
van de Loo
JWHP
Donath
MJSH
van Mourik
JA
Mertens
K
The sequence Glu1811-Lys1818 of human blood coagulation factor VIII comprises a binding site for activated factor IX.
J Biol Chem
271
1996
1935
143
Brandstetter
H
Bauer
M
Huber
R
Lollar
P
Bode
W
X-ray structure of clotting factor IXa: Active site and module structure related to Xase activity and hemophilia B.
Proc Natl Acad Sci USA
92
1995
9796
144
Pan
Y
DeFay
T
Gitschier
J
Cohen
FE
Proposed structure of the A domains of factor VIII by homology modelling.
Nature Struct Biol
2
1995
740
145
Pemberton
S
Lindley
P
Zaitsev
V
Card
G
Tuddenham
EGD
Kemball-Cook
G
A molecular model for the triplicated A domains of human factor VIII based on the crystal structure of human caeruloplasmin.
Blood
89
1997
2413
146
Bajaj
SP
Rapaport
SI
Maki
SL
A monoclonal antibody to factor IX that inhibits the factor VIII:Ca potentiation of factor X activation.
J Biol Chem
260
1985
11574
147
Bajaj
SP
Sabharwal
AK
Gorka
J
Birktoft
JJ
Antibody-probed conformational transitions in the protease domain of human factor IX upon calcium binding and zymogen activation: Putative high affinity Ca2+ binding site in the protease domain.
Proc Natl Acad Sci USA
89
1992
152
148
Hamaguchi
N
Bajaj
SP
Smith
KJ
Stafford
DW
The role of amino-terminal residues of the heavy chain of factor IXa in the binding of its cofactor, factor VIIIa.
Blood
84
1994
1837
149
Lenting
PJ
ter Maat
H
Clijsters
PPFM
Donath
MJSH
van Mourik
JA
Mertens
K
Cleavage at arginine 145 in human blood coagulation factor IX converts the zymogen into a factor VIII binding enzyme.
J Biol Chem
270
1995
14884
150
Rees
DJG
Jones
IM
Handford
PA
Walter
SJ
Esnouf
MP
Smith
KJ
Brownlee
GG
The role of β-hydroxyaspartate and adjacent carboxylate residues in the first EGF domain of human factor IX.
EMBO J
7
1988
2053
151
Hughes
PE
Morgan
G
Rooney
EK
Brownlee
GG
Handford
PA
Tyrosine 69 of the first epidermal growth factor-like domain of human factor IX is essential for clotting activity.
J Biol Chem
268
1993
17727
152
Larson
PJ
Stannfield-Oakly
S
van Dusen
WJ
Kasper
SK
Smith
KJ
Monroe
DM
High
K
Structural integrity of the gamma-carboxyglutamic acid domain of human blood coagulation factor IXa is required for its binding to cofactor VIIIa.
J Biol Chem
271
1996
3869
153
Nishimura
H
Takeya
H
Miyata
T
Suehiro
K
Okamura
T
Niho
Y
Iwanaga
S
Factor IX Fukuoka.
J Biol Chem
268
1993
24041
154
Christophe
OD
Lenting
PJ
Kolkman
JA
Rees
DJG
Mertens
K
Blood coagulation factor IX residues Glu78 and Arg94 provide a link between both epidermal growth factor-like domains that is crucial in the interaction with factor VIII light chain.
J Biol Chem
273
1998
222
155
Lenting
PJ
Christophe
OD
ter Maat
H
Rees
DJG
Mertens
K
Ca2+ binding to the first epidermal growth factor-like domain of human blood coagulation factor IX promotes enzyme activity and factor VIII light chain binding.
J Biol Chem
271
1996
25332
156
Amano
K
Sarkar
R
Pemberton
S
Kemball-Cook
G
Kazazian
HH
Kaufman
RJ
The molecular basis for cross-reacting material-positive hemophilia A due to missense mutations within the A2-domain of factor VIII.
Blood
91
1998
538
157
Mertens
K
van Wijngaarden
A
Bertina
RM
Veltkamp
JJ
The functional defect of factor VIII Leiden, a genetic variant of coagulation factor VIII.
Thromb Haemost
54
1985
650
158
Celie
PHN
Donath
MJSH
Jorieux
S
van Mourik
JA
Mazurier
C
Mertens
K
Characterization of Arg527 and Arg531 substitution variants of factor VIII associated with mild haemophilia.
Thromb Haemost
77
1997
570
(abstr, suppl)
159
Lollar
P
Knutson
GJ
Fass
DN
Stabilisation of thrombin activated porcine factor VIII:C by factor IXa and phospholipid.
Blood
63
1984
1303
160
Lollar
P
Parker
ET
Fay
PJ
Coagulant properties of hybrid human/porcine factor VIII molecules.
J Biol Chem
267
1992
23652
161
Neuenschwander
P
Jesty
J
A comparison of phospholipid and platelets in the activation of human factor VIII by thrombin and factor Xa, and in the activation of factor X.
Blood
72
1988
1761
162
Lu
D
Kalafatis
M
Mann
KG
Long
GL
Comparison of activated protein C/protein S-mediated inactivation of human factor VIII and factor V.
Blood
87
1996
4708
163
Persson
E
Ezban
M
Shymko
RM
Kinetics of the interaction between the human factor VIIIa subunits: Effects of pH, ionic strength, Ca2+ concentration, heparin, and activated protein C-catalyzed proteolysis.
Biochemistry
34
1995
12775
164
Lollar
P
Parker
CG
Subunit structure of thrombin-activated porcine factor VIII.
Biochemistry
28
1989
666
165
Lollar
P
Parker
ET
Structural basis for the decreased procoagulant activity of human factor VIII compared to the porcine homolog.
J Biol Chem
266
1991
12481
166
Fay
PJ
Haidaris
PJ
Smudzin
TM
Human factor VIIIa subunit structure.
J Biol Chem
266
1991
8957
167
Walker
FJ
Chavin
S
Fay
PJ
Inactivation of FVIII by activated protein C and protein S.
Arch Biochem Biophys
252
1987
322
168
Fay
PJ
Smudzin
TM
Walker
FJ
Activated protein C-catalyzed inactivation of human factor VIII and factor VIIIa.
J Biol Chem
266
1991
20139
169
O’Brien
DP
Johnson
D
Byfield
P
Tuddenham
EGD
Inactivation of factor VIII by factor IXa.
Biochemistry
31
1992
2805
170
Lamphear
BJ
Fay
PJ
Proteolytic interactions of factor IXa with human factor VIII and factor VIIIa.
Blood
80
1992
3120
171
Fay
PJ
Beattie
TL
Regan
LM
O’Brien
LM
Kaufman
RJ
Model for the factor VIIIa-dependent decay of the intrinsic factor Xase.
J Biol Chem
271
1996
6027
172
van’t Veer
C
Golden
NJ
Kalafatis
M
Mann
KG
Inhibitory mechanism of the protein C pathway on tissue factor-induced thrombin generation.
J Biol Chem
272
1997
7983
173
Yakhyaev
A
Mikhailenko
I
Strickland
D
Saenko
E
Cellular uptake and degradation of thrombin activated factor VIII fragments.
Blood
90
1997
31a
(abstr, suppl)
174
Bertina
RM
Cupers
R
van Wijngaarden
A
Factor IXa protects activated factor VIII against inactivation by activated protein C.
Biochem Biophys Res Commun
125
1984
177
175
Bertina
RM
Koeleman
BPC
Koster
T
Rosendaal
FR
Dirven
RJ
de Ronde
H
van der Velden
PA
Reitsma
PH
Mutation in blood coagulation factor V associated with resistance to activated protein C.
Nature
369
1994
64
176
Greengard
JS
Sun
X
Xu
X
Fernandez
JA
Griffin
JH
Evatt
B
Activated protein C resistance caused by Arg506Gln mutation in factor Va.
Lancet
343
1994
1361
177
Voorberg
J
Roelse
J
Koopman
R
Büller
H
Berends
F
ten Cate
JW
Mertens
K
van Mourik
JA
Association of idiopathic venous thromboembolism with a single point mutation at Arg506 of factor V.
Lancet
343
1994
1535
178
Zöller
B
Dählback
B
Linkage between inherited resistance to activated protein C and factor V mutation in venous thrombosis.
Lancet
343
1994
1536
179
Roelse
JC
Koopman
MMW
Büller
HR
ten Cate
JW
Montaruli
B
van Mourik
JA
Voorberg
J
Absence of mutations at the activated protein C cleavage sites of factor VIII in 125 patiens with venous thrombosis.
Br J Haematol
92
1996
740
180
Bokarewa
MI
Falk
G
Bremme
K
Blomback
M
Wiman
B
Search for mutations in the genes for coagulation factors V and VIII with a possible predisposition to activated protein C resistance.
Eur J Clin Invest
27
1997
340
181
Amano
K
Michnick
DA
Moussalli
M
Kaufman
RJ
Mutation at either Arg336 or Arg562 in factor VIII is insufficient for complete restistance to activated protein C (APC)-mediated inactivation: Implications for the APC resistance test.
Thromb Haemost
79
1998
557
182
Shen
L
Dahlbäck
B
Factor V and protein S as synergistic cofactors to activated protein C in degradation of factor VIIIa.
J Biol Chem
269
1994
18735
183
Váradi
K
Rosing
J
Tans
G
Pabinger
I
Kail
B
Schwarz
HP
Factor V enhances the cofactor function of protein S in the APC-mediated inactivation of factor VIII: Influence of the factor VR506Q mutation.
Thromb Haemost
76
1996
208
184
van ‘t Veer
C
Golden
NJ
Kalafatis
M
Simioni
P
Bertina
RM
Mann
KG
An in vitro analysis of the combination of hemophilia A and factor V-Leiden.
Blood
90
1997
3067
185
Nichols
WC
Amano
K
Cacheris
PM
Figueiredo
MS
Michaelides
K
Schwaab
R
Hoyer
L
Kaufman
RJ
Ginsburg
D
Moderation of Hemophilia A phenotype by the factor V R506Q mutation.
Blood
88
1996
1183
186
Arbini
AA
Mannucci
PM
Bauer
KA
Low prevalence of the factor V Leiden mutation among “severe” hemophiliacs with a “milder” bleeding diathesis.
Thromb Haemost
74
1995
1255
187
Tavassoli
M
Kishimoto
T
Kataoka
M
Liver endothelium mediates the hepatocytes uptake of ceruloplasmin.
J Cell Biol
102
1986
1298
188
Rand
MD
Hanson
SR
Mann
KG
Factor V turnover in a primate model.
Blood
86
1995
2616
189
Mertens
K
Leyte
A
Lenting
PJ
van Mourik
JA
Factor VIII-von Willebrand factor interaction and in vivo survival of factor VIII light chain.
Thromb Haemost
65
1991
658
(abstr)
190
Fijnvandraat
K
Berntop
E
ten Cate
JW
Johnsson
H
Peters
M
Savidge
G
Tengborn
L
Spira
J
Stahl
C
Recombinant, B-domain deleted factor VIII (r-VIII-SQ): Pharmacokinetics and initial safety aspects in hemophilia A patients.
Thromb Haemost
77
1997
298
191
Pittman
DD
Alderman
EM
Tomkinson
KN
Wang
JH
Giles
AR
Kaufman
RJ
Biochemical, immunological, and in vivo functional characterization of B-domain–deleted factor VIII.
Blood
81
1993
2925
192
Mertens
K
Donath
MJSH
van Leen
RW
de Keyzer-Nellen
MJM
Verbeet
MP
Klaasse Bos
JM
Leyte
A
van Mourik
JA
Biological activity of recombinant factor VIII variants lacking the central B-domain and the heavy chain sequence Lys713-Arg740: Discordant in vitro and in vivo activity.
Br J Haematol
85
1993
133
193
Lubin
IM
Healey
JF
Scandella
D
Runge
MS
Lollar
P
Elimenation of a major inhibitor epitope in factor VIII.
J Biol Chem
269
1994
8639
194
Healey
JF
Lubin
IM
Nakai
H
Saenko
EL
Hoyer
LW
Scandella
D
Lollar
P
Residues 484-508 contain a major determinant of the inhibitory epitope in the A2 domain of human factor VIII.
J Biol Chem
270
1995
14505
195
Pipe
SW
Kaufman
RJ
Characterization of a genetically engineered inactivation-resistant factor VIIIa.
Proc Natl Acad Sci USA
94
1997
11851
196
McMullen
BA
Fujikawa
K
Davie
EW
Hedner
U
Ezban
M
Locations of disulfide bridges and free cysteines in the heavy and light chains of recombinant human factor VIII (antihemophilic factor A).
Protein Sci
4
1995
740
197
Fay
PJ
Smudzin
TM
Inter-subunit fluorescence energy transfer in human factor VIII.
J Biol Chem
264
1989
14005
198
Sandberg
H
Brandt
J
Ålin
P
Strömberg
S
Gray
E
Bartfai
J
Castro
V
Glycosylation pattern of a B-domain-deleted factor VIII molecule (r-VIII-SQ).
Thromb Haemost
73
1995
1214
(abstr)
199
Bihoreau
N
Veillon
JF
Ramon
C
Scohyers
JM
Schmitter
JM
Characterization of a recombinant antihaemophilia A factor (factor VIII-▵II) by matrix-assisted laser desorption/ionization mass spectrometry.
Rapid Commun Mass Spectrom
9
1995
1584
200
Medzihradszky
KF
Besman
MJ
Burlingame
AL
Structural characterization of site specific N-glycosylation of recombinant human factor VIII by reversed-phase high-performance liquid chromatography-electrospray ionization mass spectrometry.
Anal Chem
69
1997
3968

Author notes

Address reprint requests to Peter J. Lenting, PhD, Department of Plasma Protein Technology, CLB, Sanquin Blood Supply Foundation, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; e-mail: P_Lenting@clb.nl.

Sign in via your Institution