Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Blood Adv ; 6(9): 2778-2790, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35015821

ABSTRACT

Type 2N von Willebrand disease is caused by mutations in the factor VIII (FVIII) binding site of von Willebrand factor (VWF), resulting in dysfunctional VWF with defective binding capacity for FVIII. We developed a novel type 2N mouse model using CRISPR/Cas9 technology. In homozygous VWF2N/2N mice, plasma VWF levels were normal (1167 ± 257 mU/mL), but the VWF was completely incapable of binding FVIII, resulting in 53 ± 23 mU/mL of plasma FVIII levels that were similar to those in VWF-deficient (VWF-/-) mice. When wild-type human or mouse VWF was infused into VWF2N/2N mice, endogenous plasma FVIII was restored, peaking at 4 to 6 hours post-infusion, demonstrating that FVIII expressed in VWF2N mice is viable but short-lived unprotected in plasma due to dysfunctional 2N VWF. The whole blood clotting time and thrombin generation were impaired in VWF2N/2N but not in VWF-/- mice. Bleeding time and blood loss in VWF2N/2N mice were similar to wild-type mice in the lateral tail vein or ventral artery injury model. However, VWF2N/2N mice, but not VWF-/- mice, lost a significant amount of blood during the primary bleeding phase after a tail tip amputation injury model, indicating that alternative pathways can at least partially restore hemostasis when VWF is absent. In summary, we have developed a novel mouse model by gene editing with both the pathophysiology and clinical phenotype found in severe type 2N patients. This unique model can be used to investigate the biological properties of VWF/FVIII association in hemostasis and beyond.


Subject(s)
Hemostatics , von Willebrand Disease, Type 2 , von Willebrand Diseases , Animals , CRISPR-Cas Systems , Disease Models, Animal , Gene Editing , Hemorrhage/genetics , Humans , Mice , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
2.
Res Pract Thromb Haemost ; 4(1): 64-71, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31989086

ABSTRACT

BACKGROUND: Von Willebrand Disease (VWD) is the most common inherited bleeding disorder, caused by quantitative and qualitative changes in von Willebrand factor (VWF). The biology of VWD, studied in canine, porcine, and murine models, differ in species-specific biology of VWF and the amenability to experimental manipulations such as phlebotomy. The factor VIII (FVIII) levels in these models are higher than in humans with type 3 VWD, suggesting functional differences between FVIII and VWF.ObjectivesTo develop a VWF knock out (VWF-/-) rat by excision of all 52 exons of the VWF locus. METHODS: The entire VWF gene was eliminated in Sprague-Dawley (Crl:SD) rats via CRISPR/Cas9-mediated gene editing. VWF antigen (VWF:Ag), VWF propeptide, and VWF collagen IV binding (VWF:CB4) levels were determined by ELISA assays and FVIII chromogenic activity (FVIII:C) levels by chromogenic FVIII assays. Lateral tail veins were transected to measure bleeding time. VWF-/- rats were infused with FVIII-/- rat platelet poor plasma (PPP) to determine response of plasma FVIII. RESULTS: Breeding of VWF ± rats yielded VWF-/- offspring at normal Mendelian ratios. VWF:Ag, VWF propeptide, VWF:CB4, and FVIII:C plasma levels were undetectable in VWF-/- rats. VWF-/- rats bled longer and more than VWF+/- and VWF+/+ rats when challenged. Transfusion of FVIII-deficient platelet-poor plasma induced a rapid rise in endogenous FVIII:C in VWF-/- rats. CONCLUSION: This rat model of severe VWD due to elimination of the entire VWF gene recapitulates the severe secondary deficiency of FVIII seen in human type 3 VWD and facilitates the study of VWF and FVIII and their interactions.

3.
Blood Adv ; 4(1): 55-65, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31899798

ABSTRACT

Previous studies have shown that platelet-specific factor VIII (FVIII) expression (2bF8) restores hemostasis and induces immune tolerance in hemophilia A (HA) mice even with preexisting inhibitors. Here we investigated for the first time whether platelet FVIII expression can prevent severe spontaneous bleeding in rat HA, a model mimicking the frequent spontaneous bleeding in patients with severe HA. A novel FVIII-/- rat model in a Dahl inbred background (Dahl-FVIII-/-) with nearly the entire rat FVIII gene inverted was created by using a CRISPR/Cas9 strategy. There was no detectable FVIII in plasma. Spontaneous bleeding in the soft tissue, muscles, or joints occurred in 100% of FVIII-/- rats. Sixty-one percent developed anti-FVIII inhibitors after ≥2 doses of recombinant human FVIII infusion. However, when 2bF8 transgene was crossed into the FVIII-/- background, none of the resulting 2bF8tg+FVIII-/- rats (with platelet FVIII levels of 28.26 ± 7.69 mU/108 platelets and undetectable plasma FVIII) ever had spontaneous bleeding. When 2bF8tg bone marrow (BM) was transplanted into FVIII-/- rats, only 1 of 7 recipients had a bruise at the early stage of BM reconstitution, but no other spontaneous bleeding was observed during the study period. To confirm that the bleeding diathesis in FVIII-/- rats was ameliorated after platelet FVIII expression, rotational thromboelastometry and whole-blood thrombin generation assay were performed. All parameters in 2bF8tg BM transplantation recipients were significantly improved compared with FVIII-/- control rats. Of note, neither detectable levels of plasma FVIII nor anti-FVIII inhibitors were detected in 2bF8tg BM transplantation recipients. Thus, platelet-specific FVIII expression can efficiently prevent severe spontaneous bleeding in FVIII-/- rats with no anti-FVIII antibody development.


Subject(s)
Factor VIII , Hemophilia A , Animals , Blood Platelets , Factor VIII/genetics , Genetic Therapy , Hemophilia A/drug therapy , Hemophilia A/genetics , Humans , Phenotype , Rats , Rats, Inbred Dahl
4.
Blood Adv ; 2(19): 2522-2532, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30287479

ABSTRACT

The interaction of platelet glycoprotein Ibα (GPIbα) with von Willebrand factor (VWF) initiates hemostasis after vascular injury and also contributes to pathological thrombosis. GPIbα binding to the VWF A1 domain (VWFA1) is a target for antithrombotic intervention, but attempts to develop pharmacologic inhibitors have been hindered by the lack of animal models because of the species specificity of the interaction. To address this problem, we generated a knockin mouse with Vwf exon 28-encoding domains A1 and A2 replaced by the human homolog (VWFh28). VWFh28 mice (M1HA) were crossbred with a transgenic mouse strain expressing human GPIbα on platelets (mGPIbαnull;hGPIbαTg; H1MA) to generate a new strain (H1HA) with humanized GPIbα-VWFA1 binding. Plasma VWF levels in the latter 3 strains were similar to those of wild-type mice (M1MA). Compared with the strains that had homospecific GPIbα-VWF pairing (M1MA and H1HA), M1HA mice of those with heterospecific pairing had a markedly greater prolongation of tail bleeding time and attenuation of thrombogenesis after injury to the carotid artery than H1MA mice. Measurements of GPIbα-VWFA1 binding affinity by surface plasmon resonance agreed with the extent of observed functional defects. Ristocetin-induced platelet aggregation was similar in H1HA mouse and human platelet-rich plasma, and it was comparably inhibited by monoclonal antibody NMC-4, which is known to block human GPIbα-VWFA1 binding, which also inhibited FeCl3-induced mouse carotid artery thrombosis. Thus, the H1HA mouse strain is a fully humanized model of platelet GPIbα-VWFA1 binding that provides mechanistic and pharmacologic information relevant to human hemostatic and thrombotic disorders.


Subject(s)
Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism , Animals , Biomarkers , Blood Platelets/metabolism , Crosses, Genetic , Exons , Hemostasis , Humans , Mice , Mice, Transgenic , Molecular Docking Simulation , Molecular Dynamics Simulation , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Aggregates , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship , Surface Plasmon Resonance , Thrombosis/etiology , Thrombosis/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/genetics
5.
Blood ; 123(24): 3706-13, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24705491

ABSTRACT

The cellular source of coagulation factor VIII (FVIII) remains controversial. Like many coagulation proteins, FVIII is produced in the liver, and FVIII synthesis has long been associated with hepatocytes. But extrahepatic synthesis also occurs, and mounting evidence suggests that hepatocytes are not responsible for FVIII production. To determine the tissue that synthesizes FVIII, we developed a Cre/lox-dependent conditional knockout (KO) model in which exons 17 and 18 of the murine factor VIII gene (F8) are flanked by loxP sites, or floxed (F8(F)). In cells expressing Cre-recombinase, the floxed sequence is deleted, resulting in F8(F→KO) gene inactivation. When F8(F) mice were crossed with various tissue-specific Cre strains, we found that hepatocyte-specific F8-KO mice are indistinguishable from controls, whereas efficient endothelial-KO models display a severe hemophilic phenotype with no detectable plasma FVIII activity. A hematopoietic Cre model was more equivocal, so experimental bone marrow transplantation was used to examine hematopoietic FVIII synthesis. FVIII(null) mice that received bone marrow transplants from wild-type donors were still devoid of plasma FVIII activity after hematopoietic donor cell engraftment. Our results indicate that endothelial cells are the predominant, and possibly exclusive, source of plasma FVIII.


Subject(s)
Endothelial Cells/metabolism , Factor VIII/biosynthesis , Animals , Bone Marrow Transplantation , Cells, Cultured , Factor VIII/analysis , Factor VIII/genetics , Female , Genotype , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tissue Distribution
6.
Blood ; 123(3): 395-403, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24269957

ABSTRACT

Our previous studies have demonstrated that platelet FVIII (2bF8) gene therapy can improve hemostasis in hemophilia A mice, even in the presence of inhibitory antibodies, but none of our studies has targeted human cells. Here, we evaluated the feasibility for lentivirus (LV)-mediated human platelet gene therapy of hemophilia A. Human platelet FVIII expression was introduced by 2bF8LV-mediated transduction of human cord blood (hCB) CD34(+) cells followed by xenotransplantation into immunocompromised NSG mice or NSG mice in an FVIII(null) background (NSGF8KO). Platelet FVIII was detected in all recipients that received 2bF8LV-transduced hCB cells as long as human platelet chimerism persisted. All NSGF8KO recipients (n = 7) that received 2bF8LV-transduced hCB cells survived tail clipping if animals had greater than 2% of platelets derived from 2bF8LV-transduced hCB cells, whereas 5 of 7 survived when human platelets were 0.3% to 2%. Whole blood clotting time analysis confirmed that hemostasis was improved in NSGF8KO mice that received 2bF8LV-transduced hCB cells. We demonstrate, for the first time, the feasibility of 2bF8LV gene delivery to human hematopoietic stem cells to introduce FVIII expression in human platelets and that human platelet-derived FVIII can improve hemostasis in hemophilia A.


Subject(s)
Blood Platelets/metabolism , Cord Blood Stem Cell Transplantation , Gene Expression Regulation , Genetic Therapy , Hemophilia A/genetics , Hemophilia A/therapy , Animals , Antigens, CD34/metabolism , Blood Platelets/cytology , Chimerism , Factor VIII/metabolism , Fetal Blood/metabolism , Humans , Immunohistochemistry , Lentivirus/genetics , Lentivirus/metabolism , Mice , Mice, Knockout , Mice, SCID , Phenotype , Thrombopoiesis
7.
Mol Ther ; 20(3): 625-32, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22044935

ABSTRACT

Bernard-Soulier syndrome (BSS) is an inherited bleeding disorder caused by a defect in the platelet glycoprotein (GP) Ib-IX-V complex. The main treatment for BSS is platelet transfusion but it is often limited to severe bleeding episodes or surgical interventions due to the risk of alloimmunization. We have previously reported successful expression of human GPIbα (hGPIbα) in human megakaryocytes using a lentiviral vector (LV) encoding human GP1BA under control of the platelet-specific integrin αIIb promoter (2bIbα). In this study, we examined the efficacy of this strategy for the gene therapy of BSS using GPIbα(null) as a murine model of BSS. GPIbα(null) hematopoietic stem cells (HSC) transduced with 2bIbα LV were transplanted into lethally irradiated GPIbα(null) littermates. Therapeutic levels of hGPIbα expression were achieved that corrected the tail bleeding time and improved the macrothrombocytopenia. Sequential bone marrow (BM) transplants showed sustained expression of hGPIbα with similar phenotypic correction. Antibody response to hGPIbα was documented in 1 of 17 total recipient mice but was tolerated without any further treatment. These results demonstrate that lentivirus-mediated gene transfer can provide sustained phenotypic correction of murine BSS, indicating that this approach may be a promising strategy for gene therapy of BSS patients.


Subject(s)
Bernard-Soulier Syndrome/therapy , Genetic Therapy , Genetic Vectors/genetics , Lentivirus/genetics , Animals , Antibodies/blood , Antibodies/immunology , Bernard-Soulier Syndrome/genetics , Bernard-Soulier Syndrome/immunology , Blood Platelets/metabolism , Bone Marrow Transplantation , Disease Models, Animal , Gene Expression , Hematopoietic Stem Cells/metabolism , Hemorrhage/genetics , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Glycoprotein GPIb-IX Complex , Protein Binding , Thrombocytopenia/immunology
8.
Blood ; 116(16): 3049-57, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20606161

ABSTRACT

The natural cell type(s) that synthesize and release factor VIII (FVIII) into the circulation are still not known with certainty. In vitro studies indicate that artificial expression of FVIII in endothelial cells produces an intracellular pool of FVIII that can be mobilized together with its carrier protein, von Willebrand factor (VWF), by agonists. Here, we show that expression of human B-domain deleted FVIII (hFVIII) in the vascular endothelium of otherwise FVIII-deficient mice results in costorage of FVIII and VWF in endothelial Weibel-Palade bodies and restores normal levels and activity of FVIII in plasma. Stored FVIII was mobilized into the circulation by subcutaneous administration of epinephrine. Human FVIII activity in plasma was strictly dependent on the presence of VWF. Endothelial-specific expression of hFVIII rescued the bleeding diathesis of hemophilic mice lacking endogenous FVIII. This hemostatic function of endothelial cell-derived hFVIII was suppressed in the presence of anti-FVIII inhibitory antibodies. These results suggest that targeting FVIII expression to endothelial cells may establish a releasable pool of FVIII and normalize plasma FVIII level and activity in hemophilia A, but does not prevent the inhibitory effect of anti-FVIII antibodies on the hemostatic function of transgene-derived hFVIII as is seen with platelet-derived FVIII expression.


Subject(s)
Endothelium, Vascular/metabolism , Factor VIII/metabolism , Hemophilia A/metabolism , Hemostasis , Weibel-Palade Bodies/metabolism , von Willebrand Factor/metabolism , Animals , Antibodies/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Epinephrine/pharmacology , Factor VIII/genetics , Factor VIII/immunology , Gene Expression , Humans , Mice , Mice, Transgenic , Protein Structure, Tertiary
9.
Blood ; 116(8): 1235-43, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20445020

ABSTRACT

We developed 2bF9 transgenic mice in a hemophilia B mouse model with the expression of human factor IX (FIX) under control of the platelet-specific integrin alphaIIb promoter, to determine whether ectopically expressing FIX in megakaryocytes can enable the storage of FIX in platelet alpha-granules and corrects the murine hemophilia B phenotype. FIX was detected in the platelets and plasma of 2bF9 transgenic mice by both antigen and activity assays. Approximately 90% of total FIX in blood was stored in platelets, most of which is releasable on activation of platelets. Immunostaining demonstrated that FIX was expressed in platelets and megakaryocytes and stored in alpha-granules. All 2bF9 transgenic mice survived tail clipping, suggesting that platelet-derived FIX normalizes hemostasis in the hemophilia B mouse model. This protection can be transferred by bone marrow transplantation or platelet transfusion. However, unlike our experience with platelet FVIII, the efficacy of platelet-derived FIX was limited in the presence of anti-FIX inhibitory antibodies. These results demonstrate that releasable FIX can be expressed and stored in platelet alpha-granules and that platelet-derived FIX can correct the bleeding phenotype in hemophilia B mice. Our studies suggest that targeting FIX expression to platelets could be a new gene therapy strategy for hemophilia B.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Granules/metabolism , Factor IX/metabolism , Genetic Therapy , Hemophilia B/therapy , Animals , Blood Coagulation , Blotting, Western , Bone Marrow Transplantation , Factor IX/genetics , Factor VIII/genetics , Factor VIII/metabolism , Female , Hemophilia B/genetics , Hemophilia B/pathology , Humans , Immunization , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Platelet Count , Platelet Membrane Glycoprotein IIb/genetics , Promoter Regions, Genetic/genetics
10.
Blood ; 112(7): 2713-21, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18495954

ABSTRACT

Although genetic induction of factor VIII (FVIII) expression in platelets can restore hemostasis in hemophilia A mice, this approach has not been studied in the clinical setting of preexisting FVIII inhibitory antibodies to determine whether such antibodies would affect therapeutic engraftment. We generated a line of transgenic mice (2bF8) that express FVIII only in platelets using the platelet-specific alphaIIb promoter and bred this 2bF8 transgene into a FVIII(null) background. Bone marrow (BM) from heterozygous 2bF8 transgenic (2bF8(tg+/-)) mice was transplanted into immunized FVIII(null) mice after lethal or sublethal irradiation. After BM reconstitution, 85% of recipients survived tail clipping when the 1100-cGy (myeloablative) regimen was used, 85.7% of recipients survived when 660-cGy (nonmyeloablative) regimens were used, and 60% of recipients survived when the recipients were conditioned with 440 cGy. Our further studies showed that transplantation with 1% to 5% 2bF8(tg+/-) BM cells still improved hemostasis in hemophilia A mice with inhibitors. These results demonstrate that the presence of FVIII-specific immunity in recipients does not negate engraftment of 2bF8 genetically modified hematopoietic stem cells, and transplantation of these hematopoietic stem cells can efficiently restore hemostasis to hemophilic mice with preexisting inhibitory antibodies under either myeloablative or nonmyeloablative regimens.


Subject(s)
Blood Platelets/metabolism , Factor VIII/therapeutic use , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hemophilia A/genetics , Hemostasis , Immunity , Animals , Biological Assay , Blood Platelets/drug effects , Factor VIII/genetics , Factor VIII/immunology , Genetic Therapy , Hematopoietic Stem Cells/drug effects , Hemophilia A/therapy , Immunization , Mice , Myeloablative Agonists/pharmacology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transformation, Genetic , Transgenes , Transplantation Conditioning , Transplantation, Isogeneic
11.
J Clin Invest ; 116(7): 1974-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16823491

ABSTRACT

Inhibitory immune response to exogenously infused factor VIII (FVIII) is a major complication in the treatment of hemophilia A. Generation of such inhibitors has the potential to disrupt gene therapy for hemophilia A. We explore what we believe to be a novel approach to overcome this shortcoming. Human B-domain-deleted FVIII (hBDDFVIII) was expressed under the control of the platelet-specific alphaIIb promoter in platelets of hemophilic (FVIIInull) mice to create 2bF8trans mice. The FVIII transgene product was stored in platelets and released at the site of platelet activation. In spite of the lack of FVIII in the plasma of 2bF8trans mice, the bleeding phenotype of FVIIInull mice was corrected. More importantly, the bleeding phenotype was corrected in the presence of high inhibitory antibody titers introduced into the mice by infusion or by spleen cell transfer from recombinant hBDDFVIII-immunized mice. Our results demonstrate that this approach to the targeted expression of FVIII in platelets has the potential to correct hemophilia A, even in the presence of inhibitory immune responses to infused FVIII.


Subject(s)
Antibodies/immunology , Blood Platelets/physiology , Factor VIII/metabolism , Factor VIII/therapeutic use , Hemophilia A , Animals , Factor VIII/genetics , Genetic Therapy , Hemophilia A/genetics , Hemophilia A/immunology , Hemophilia A/therapy , Hemostasis/physiology , Humans , Mice , Mice, Transgenic , Phenotype , Platelet Activation , Transgenes , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
12.
Blood ; 105(1): 145-52, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15331450

ABSTRACT

Type 3 von Willebrand disease (VWD) is a severe hemorrhagic defect in humans. We now identify the homozygous mutation in the Chapel Hill strain of canine type 3 VWD that results in premature termination of von Willebrand factor (VWF) protein synthesis. We cultured endothelium from VWD and normal dogs to study intracellular VWF trafficking and Weibel-Palade body formation. Weibel-Palade bodies could not be identified in the canine VWD aortic endothelial cells (VWD-AECs) by P-selectin, VWFpp, or VWF immunostaining and confocal microscopy. We demonstrate the reestablishment of Weibel-Palade bodies that recruit endogenous P-selectin by expressing wild-type VWF in VWD-AECs. Expression of mutant VWF proteins confirmed that VWF multimerization is not necessary for Weibel-Palade body creation. Although the VWF propeptide is required for the formation of Weibel-Palade bodies, it cannot independently induce the formation of the granule. These VWF-null endothelial cells provide a unique opportunity to examine the biogenesis of Weibel-Palade bodies in endothelium from a canine model of type 3 VWD.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Weibel-Palade Bodies/metabolism , von Willebrand Diseases/metabolism , von Willebrand Diseases/pathology , von Willebrand Factor/metabolism , Amino Acid Sequence , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Base Sequence , Cells, Cultured , Dogs , Frameshift Mutation/genetics , Gene Expression Regulation , Humans , Molecular Sequence Data , Protein Processing, Post-Translational , Weibel-Palade Bodies/genetics , Weibel-Palade Bodies/pathology , von Willebrand Diseases/genetics , von Willebrand Factor/chemistry , von Willebrand Factor/genetics
13.
Blood ; 102(2): 436-41, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12649145

ABSTRACT

Recombinant human interleukin-11 (rhIL-11), a glycoprotein 130 (gp130)-signaling cytokine approved for treatment of thrombocytopenia, also raises von Willebrand factor (VWF) and factor VIII (FVIII) by an unknown mechanism. Desmopressin (1-deamino-8-d-arginine vasopressin [DDAVP]) releases stored VWF and FVIII and is used for treatment of VWF and FVIII deficiencies. To compare the effect of these 2 agents, heterozygous von Willebrand disease (VWD) and normal dogs were treated with either rhIL-11 (50 microg/kg/d subcutaneously x 7 days) or DDAVP (5 microg/kg/d intravenously x 7 days). The rhIL-11 produced a gradual and sustained elevation of VWF and FVIII levels in both heterozygous VWD and normal dogs while DDAVP produced a rapid and unsustained increase. Importantly, rhIL-11 treatment produced a 2.5- to 11-fold increase in VWF mRNA in normal canine heart, aorta, and spleen but not in homozygous VWD dogs, thus identifying a mechanism for elevation of plasma VWF in vivo. Moreover, dogs pretreated with rhIL-11 retain a DDAVP-releasable pool of VWF and FVIII, suggesting that rhIL-11 does not significantly alter trafficking of these proteins to or from storage pools. The half-life of infused VWF is unchanged by rhIL-11 in homozygous VWD dogs. These results show that rhIL-11 and DDAVP raise plasma VWF by different mechanisms. Treatment with rhIL-11 with or without DDAVP may provide an alternative to plasma-derived products for some VWD and hemophilia A patients if it is shown safe in clinical trials.


Subject(s)
Deamino Arginine Vasopressin/therapeutic use , Interleukin-11/therapeutic use , RNA, Messenger/biosynthesis , Weibel-Palade Bodies/metabolism , von Willebrand Diseases/physiopathology , von Willebrand Factor/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Deamino Arginine Vasopressin/pharmacology , Dogs , Drug Evaluation, Preclinical , Factor VIII/metabolism , Half-Life , Heart/drug effects , Heterozygote , Interleukin-11/pharmacology , Myocardium/metabolism , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Spleen/drug effects , Spleen/metabolism , Weibel-Palade Bodies/drug effects , von Willebrand Diseases/drug therapy , von Willebrand Diseases/genetics , von Willebrand Factor/biosynthesis , von Willebrand Factor/genetics
14.
Blood ; 100(5): 1699-706, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12176890

ABSTRACT

While studying patient plasma containing an unusual pattern of von Willebrand factor (VWF) multimers, we discovered a previously unreported phenomenon: heavy predominance of dimeric VWF. Genomic analysis revealed a new congenital mutation (Tyr87Ser) that altered the final stages of VWF biosynthesis. This mutation in the propeptide (VWFpp) resulted in synthesis of dimeric VWF with an almost complete loss of N-terminal multimerization. The multimer pattern in patient plasma appears to result from separate alleles' synthesizing wild-type or mutant (dimeric) VWF, with homodimers composing the predominant protomeric species. We have expressed VWF protein containing the Tyr87Ser mutation and analyzed the intracellular processing and resulting VWF biological functions. The expressed dimeric VWF displayed a loss of several specific functions: collagen binding, factor VIII binding, and ristocetin-induced platelet binding. However, granular storage of dimeric VWF was normal, demonstrating that the lack of multimerization does not preclude granular storage. Although the tertiary structure of the VWFpp remains unknown, the mutant amino acid is located in a region that is highly conserved across several species and may play a major role in the multimerization of VWF. Our data suggest that one function of the highly cysteine-rich VWFpp is to align the adjacent subunits of VWF into the correct configuration, serving as an intramolecular chaperone. The integrity of the VWFpp is essential to maintain the proper spacing and alignment of the multiple cysteines in the VWFpp and N-terminus of the mature VWF.


Subject(s)
von Willebrand Factor/chemistry , Amino Acid Sequence , Blood Platelets/chemistry , Blood Platelets/metabolism , Dimerization , Humans , Molecular Sequence Data , Protein Binding , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Structure, Tertiary/genetics , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...