Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Blood Adv ; 6(18): 5415-5428, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35736672

ABSTRACT

Stored red blood cells (RBCs) incur biochemical and morphological changes, collectively termed the storage lesion. Functionally, the storage lesion manifests as slower oxygen unloading from RBCs, which may compromise the efficacy of transfusions where the clinical imperative is to rapidly boost oxygen delivery to tissues. Recent analysis of large real-world data linked longer storage with increased recipient mortality. Biochemical rejuvenation with a formulation of adenosine, inosine, and pyruvate can restore gas-handling properties, but its implementation is impractical for most clinical scenarios. We tested whether storage under hypoxia, previously shown to slow biochemical degradation, also preserves gas-handling properties of RBCs. A microfluidic chamber, designed to rapidly switch between oxygenated and anoxic superfusates, was used for single-cell oxygen saturation imaging on samples stored for up to 49 days. Aliquots were also analyzed flow cytometrically for side-scatter (a proposed proxy of O2 unloading kinetics), metabolomics, lipidomics, and redox proteomics. For benchmarking, units were biochemically rejuvenated at 4 weeks of standard storage. Hypoxic storage hastened O2 unloading in units stored to 35 days, an effect that correlated with side-scatter but was not linked to posttranslational modifications of hemoglobin. Although hypoxic storage and rejuvenation produced distinct biochemical changes, a subset of metabolites including pyruvate, sedoheptulose 1-phosphate, and 2/3 phospho-d-glycerate, was a common signature that correlated with changes in O2 unloading. Correlations between gas handling and lipidomic changes were modest. Thus, hypoxic storage of RBCs preserves key metabolic pathways and O2 exchange properties, thereby improving the functional quality of blood products and potentially influencing transfusion outcomes.


Subject(s)
Blood Preservation , Oxygen , Adenosine/metabolism , Blood Preservation/methods , Erythrocytes/metabolism , Hemoglobins/metabolism , Humans , Hypoxia/metabolism , Inosine/metabolism , Oxygen/metabolism , Phosphates/metabolism , Pyruvates/metabolism
3.
Transfusion ; 62(1): 173-182, 2022 01.
Article in English | MEDLINE | ID: mdl-34757639

ABSTRACT

BACKGROUND: The COVID19 pandemic highlights the need for contingency planning in the event of blood shortages. To increase platelet supply, we assessed the operational impact and effect on platelet quality of splitting units prior to storage. STUDY DESIGN AND METHODS: Using production figures, we modeled the impact on unit numbers, platelet counts, and volumes of splitting only apheresis double donations into three units (yielding ⅔ doses), or all standard dose units in half. To assess quality, eight pools of three ABO/Rh-matched apheresis (Trima Accel) double donations in plasma were split to ⅔ and ½ volumes in both Terumo and Fresenius storage bags. These were irradiated and subject to maximal permitted periods of nonagitation (3 × 8 h) before comparing platelet quality markers (including pH, CD62P expression) to Day 9 of storage. RESULTS: Splitting all double donations into three predicted inventory expansion of 23% overall whereas halving all standard dose units clearly doubles stock. In our study, ⅔ and ½ doses contained 153 ± 15 × 109 (~138 ml) and 113 ± 11 × 109 (~102 ml) platelets respectively. Following storage, higher pH was observed in ⅔ than in ½ doses and in Terumo compared to Fresenius bags. The higher pH was reflected in better quality markers, including lower CD62P expression. Despite the differences, on Day 8 (of pH monitoring at expiry) all ⅔ doses and most ½ doses were ≥pH 6.4. CONCLUSION: A strategy to split apheresis platelets in plasma to lower doses is feasible, maintains acceptable platelet quality, and should be considered by blood services in response to extreme shortages.


Subject(s)
Blood Platelets , COVID-19 , Blood Platelets/metabolism , Blood Preservation , Humans , Platelet Count , Plateletpheresis
4.
Transfusion ; 59(9): 2952-2963, 2019 09.
Article in English | MEDLINE | ID: mdl-31294868

ABSTRACT

BACKGROUND: Rejuvenation of stored red blood cells (RBCs) increases levels of adenosine 5'-triphosphate (ATP) and 2,3-diphosphoglycerate (2,3-DPG) to those of fresh cells. This study aimed to optimize and validate the US-approved process to a UK setting for manufacture and issue of rejuvenated RBCs for a multicenter randomized controlled clinical trial in cardiac surgery. STUDY DESIGN AND METHODS: Rejuvenation of leukoreduced RBC units involved adding a solution containing pyruvate, inosine, phosphate, and adenine (Rejuvesol, Zimmer Biomet), warming at 37°C for 60 minutes, then "manual" washing with saline adenine glucose mannitol solution. A laboratory study was conducted on six pools of ABO/D-matched units made the day after donation. On Days 7, 21, and 28 of 4 ± 2°C storage, one unit per pool was rejuvenated and measured over 96 hours for volume, hematocrit, hemolysis, ATP, 2,3-DPG, supernatant potassium, lactate, and purines added (inosine) or produced (hypoxanthine) by rejuvenation. Subsequently, an operational validation (two phases of 32 units each) was undertaken, with results from the first informing a trial component specification applied to the second. Rejuvenation effects were also tested on crossmatch reactivity and RBC antigen profiles. RESULTS: Rejuvenation raised 2,3-DPG to, and ATP above, levels of fresh cells. The final component had potassium and hemolysis values below those of standard storage Days 7 and 21, respectively, containing 1.2% exogenous inosine and 500 to 1900 µmoles/unit of hypoxanthine. The second operational validation met compliance to the trial component specification. Rejuvenation did not adversely affect crossmatch reactivity or RBC antigen profiles. CONCLUSION: The validated rejuvenation process operates within defined quality limits, preserving RBC immunophenotypes, enabling manufacture for clinical trials.


Subject(s)
Blood Preservation/methods , Erythrocytes/physiology , Regenerative Medicine/methods , Rejuvenation/physiology , 2,3-Diphosphoglycerate/metabolism , Adenosine Triphosphate/blood , Blood Grouping and Crossmatching , Blood Loss, Surgical/prevention & control , Blood Preservation/standards , Cardiac Surgical Procedures/adverse effects , Cryopreservation/methods , Erythrocyte Count , Erythrocyte Transfusion/standards , Erythrocytes/cytology , Hemolysis/physiology , Humans , Immunophenotyping , Manufactured Materials , Purines/blood , Quality Control , Randomized Controlled Trials as Topic , Regenerative Medicine/standards
5.
Blood ; 131(9): 1000-1011, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29187380

ABSTRACT

Mutations in NBEAL2, the gene encoding the scaffolding protein Nbeal2, are causal of gray platelet syndrome (GPS), a rare recessive bleeding disorder characterized by platelets lacking α-granules and progressive marrow fibrosis. We present here the interactome of Nbeal2 with additional validation by reverse immunoprecipitation of Dock7, Sec16a, and Vac14 as interactors of Nbeal2. We show that GPS-causing mutations in its BEACH domain have profound and possible effects on the interaction with Dock7 and Vac14, respectively. Proximity ligation assays show that these 2 proteins are physically proximal to Nbeal2 in human megakaryocytes. In addition, we demonstrate that Nbeal2 is primarily localized in the cytoplasm and Dock7 on the membrane of or in α-granules. Interestingly, platelets from GPS cases and Nbeal2-/- mice are almost devoid of Dock7, resulting in a profound dysregulation of its signaling pathway, leading to defective actin polymerization, platelet activation, and shape change. This study shows for the first time proteins interacting with Nbeal2 and points to the dysregulation of the canonical signaling pathway of Dock7 as a possible cause of the aberrant formation of platelets in GPS cases and Nbeal2-deficient mice.


Subject(s)
Blood Platelets/metabolism , Blood Proteins/metabolism , GTPase-Activating Proteins/metabolism , Megakaryocytes/metabolism , Membrane Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Blood Platelets/cytology , Blood Proteins/genetics , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Megakaryocytes/cytology , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Protein Binding , Vesicular Transport Proteins/genetics
6.
Br J Haematol ; 175(3): 381-392, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27650431

ABSTRACT

Allogeneic platelet transfusions are widely used for the prevention and treatment of bleeding in thrombocytopenia. Recent evidence suggests platelet transfusions have limited efficacy and are associated with uncertain immunomodulatory risks and concerns about viral or bacterial transmission. Alternatives to transfusion are a well-recognised tenet of Patient Blood Management, but there has been less focus on different strategies to reduce bleeding risk by comparison to platelet transfusion. Direct alternatives to platelet transfusion include agents to stimulate endogenous platelet production (thrombopoietin mimetics), optimising platelet adhesion to endothelium by treating anaemia or increasing von Willebrand factor levels (desmopressin), increasing formation of cross-linked fibrinogen (activated recombinant factor VII, fibrinogen concentrate or recombinant factor XIII), decreasing fibrinolysis (tranexamic acid or epsilon aminocaproic acid) or using artificial or modified platelets (cryopreserved platelets, lyophilised platelets, haemostatic particles, liposomes, engineered nanoparticles or infusible platelet membranes). The evidence base to support the use of these alternatives is variable, but an area of active research. Much of the current randomised controlled trial focus is on evaluation of the use of thrombopoietin mimetics and anti-fibrinolytics. It is also recognised that one alternative strategy to platelet transfusion is choosing not to transfuse at all.


Subject(s)
Complementary Therapies , Hemorrhage/prevention & control , Hemorrhage/therapy , Platelet Transfusion , Thrombocytopenia/complications , Antifibrinolytic Agents/therapeutic use , Biological Mimicry , Blood Coagulation Factors/therapeutic use , Blood Transfusion , Complementary Therapies/methods , Humans , Nanoparticles , Platelet Transfusion/adverse effects , Thrombopoietin/therapeutic use
7.
Platelets ; 27(6): 526-34, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27135140

ABSTRACT

A goal of platelet storage is to maintain the quality of platelets from the point of donation to the point of transfusion - to suspend the aging process. This effort is judged by clinical and laboratory measures with varying degrees of success. Recent work gives encouragement that platelets can be maintained ex vivo beyond the current 5 -7 day shelf life whilst maintaining their quality, as measured by posttransfusion recovery and survival. However, additional measures are needed to validate the development of technologies that may further reduce the aging of stored platelets, or enhance their hemostatic properties.


Subject(s)
Blood Platelets/physiology , Blood Preservation , Cellular Senescence , Platelet Transfusion , Animals , Blood Preservation/adverse effects , Blood Safety , Humans , Platelet Transfusion/adverse effects , Platelet Transfusion/methods , Platelet Transfusion/standards , Platelet-Rich Plasma , Treatment Outcome
8.
Transfusion ; 54(6): 1478-85, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24308361

ABSTRACT

BACKGROUND: Studies show that 1 in 1200 neonates have a low platelet (PLT) count due to alloimmunization against human PLT antigen (HPA)-1a (ß3 -L33). This mainly occurs in HPA-1a-negative mothers who are positive for the human leukocyte antigen (HLA)-DRB3*01:01 allele, but only about one-third of cases will mount an effective alloimmune response. The development of specific treatment modalities requires that the mechanisms driving the maternal alloimmune response against the fetal PLTs be further explored. An antibody reagent that has a different binding affinity to HLA-DRA/DRB3*01:01 with and without the ß3 -L33 peptide would be a valuable reagent to study peptide presentation on maternal antigen-presenting cells. STUDY DESIGN AND METHODS: To identify such antibodies, HLA-DRA/DRB3*01:01 was recombinantly expressed in Drosophila S2 cells. To delineate the epitope of interesting antibodies, seven mutant HLA-DRA/DRB3*01:01 molecules were generated by site-directed mutagenesis introducing naturally occurring amino acid changes encoded by DRB3*02 and DRB3*03 alleles. RESULTS: The murine monoclonal antibody (MoAb) DA2 showed robust binding by enzyme-linked immunosorbent assay to recombinant HLA-DRA/DRB3*01:01, but binding was reduced in the presence of ß3 -L33 peptide. The binding affinity of DA2 to the mutant HLA-DRA/DRB3*0101 in which serine at Position 60 of the ß1-chain was replaced by tyrosine was greatly enhanced. Interestingly the binding of DA2 to the mutant was not reduced by the presence of ß3 -L33 peptide. CONCLUSION: The results of this study generate a molecular model of the interaction of the HLA-DRA/DRB3*01:01 molecule with MoAb DA2. This will inform functional studies with the recombinant Class II molecules.


Subject(s)
Antibodies, Monoclonal/metabolism , HLA Antigens/metabolism , HLA-DR alpha-Chains/metabolism , HLA-DRB3 Chains/metabolism , Antigens, Human Platelet/metabolism , Binding Sites , Enzyme-Linked Immunosorbent Assay , HLA-DR alpha-Chains/chemistry , HLA-DRB3 Chains/chemistry , Humans , Integrin beta3 , Protein Binding , Protein Structure, Secondary
9.
Nat Genet ; 45(5): 542-545, 2013 May.
Article in English | MEDLINE | ID: mdl-23563608

ABSTRACT

The blood group Vel was discovered 60 years ago, but the underlying gene is unknown. Individuals negative for the Vel antigen are rare and are required for the safe transfusion of patients with antibodies to Vel. To identify the responsible gene, we sequenced the exomes of five individuals negative for the Vel antigen and found that four were homozygous and one was heterozygous for a low-frequency 17-nucleotide frameshift deletion in the gene encoding the 78-amino-acid transmembrane protein SMIM1. A follow-up study showing that 59 of 64 Vel-negative individuals were homozygous for the same deletion and expression of the Vel antigen on SMIM1-transfected cells confirm SMIM1 as the gene underlying the Vel blood group. An expression quantitative trait locus (eQTL), the common SNP rs1175550 contributes to variable expression of the Vel antigen (P = 0.003) and influences the mean hemoglobin concentration of red blood cells (RBCs; P = 8.6 × 10(-15)). In vivo, zebrafish with smim1 knockdown showed a mild reduction in the number of RBCs, identifying SMIM1 as a new regulator of RBC formation. Our findings are of immediate relevance, as the homozygous presence of the deletion allows the unequivocal identification of Vel-negative blood donors.


Subject(s)
Blood Group Antigens/genetics , Erythrocyte Membrane/metabolism , Erythrocytes/immunology , Gene Deletion , Homozygote , Membrane Proteins/genetics , Quantitative Trait Loci , Alleles , Animals , Biomarkers/metabolism , Blood Group Antigens/immunology , Blood Group Antigens/metabolism , Electrophoretic Mobility Shift Assay , Erythrocytes/metabolism , Erythrocytes/pathology , Exome/genetics , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Isoantibodies/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pregnancy , Zebrafish/genetics
10.
Blood ; 120(24): 4859-68, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-22972982

ABSTRACT

We recently identified 68 genomic loci where common sequence variants are associated with platelet count and volume. Platelets are formed in the bone marrow by megakaryocytes, which are derived from hematopoietic stem cells by a process mainly controlled by transcription factors. The homeobox transcription factor MEIS1 is uniquely transcribed in megakaryocytes and not in the other lineage-committed blood cells. By ChIP-seq, we show that 5 of the 68 loci pinpoint a MEIS1 binding event within a group of 252 MK-overexpressed genes. In one such locus in DNM3, regulating platelet volume, the MEIS1 binding site falls within a region acting as an alternative promoter that is solely used in megakaryocytes, where allelic variation dictates different levels of a shorter transcript. The importance of dynamin activity to the latter stages of thrombopoiesis was confirmed by the observation that the inhibitor Dynasore reduced murine proplatelet for-mation in vitro.


Subject(s)
Blood Platelets/metabolism , Dynamin III/genetics , Genome, Human/genetics , Homeodomain Proteins/genetics , Megakaryocytes/metabolism , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Binding Sites/genetics , Blood Platelets/drug effects , Cell Line, Tumor , Cell Lineage/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression , Genetic Variation , Homeodomain Proteins/metabolism , Humans , Hydrazones/pharmacology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , Platelet Count , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Transcription Initiation Site , Transcription, Genetic
11.
Nat Genet ; 44(4): 435-9, S1-2, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22366785

ABSTRACT

The exon-junction complex (EJC) performs essential RNA processing tasks. Here, we describe the first human disorder, thrombocytopenia with absent radii (TAR), caused by deficiency in one of the four EJC subunits. Compound inheritance of a rare null allele and one of two low-frequency SNPs in the regulatory regions of RBM8A, encoding the Y14 subunit of EJC, causes TAR. We found that this inheritance mechanism explained 53 of 55 cases (P < 5 × 10(-228)) of the rare congenital malformation syndrome. Of the 53 cases with this inheritance pattern, 51 carried a submicroscopic deletion of 1q21.1 that has previously been associated with TAR, and two carried a truncation or frameshift null mutation in RBM8A. We show that the two regulatory SNPs result in diminished RBM8A transcription in vitro and that Y14 expression is reduced in platelets from individuals with TAR. Our data implicate Y14 insufficiency and, presumably, an EJC defect as the cause of TAR syndrome.


Subject(s)
Genetic Predisposition to Disease , RNA-Binding Proteins/genetics , Thrombocytopenia/genetics , Upper Extremity Deformities, Congenital/genetics , 5' Untranslated Regions/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes , Female , Genetic Variation , Humans , Infant , Infant, Newborn , Male , Mutation , Platelet Count , Polymorphism, Single Nucleotide , Radius/abnormalities , Sequence Alignment , Sequence Analysis, DNA , Thrombocytopenia/congenital , Young Adult , Zebrafish/genetics
12.
Nat Genet ; 43(8): 735-7, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21765411

ABSTRACT

Gray platelet syndrome (GPS) is a predominantly recessive platelet disorder that is characterized by mild thrombocytopenia with large platelets and a paucity of α-granules; these abnormalities cause mostly moderate but in rare cases severe bleeding. We sequenced the exomes of four unrelated individuals and identified NBEAL2 as the causative gene; it has no previously known function but is a member of a gene family that is involved in granule development. Silencing of nbeal2 in zebrafish abrogated thrombocyte formation.


Subject(s)
Blood Platelets/metabolism , Cytoplasmic Granules/metabolism , Gray Platelet Syndrome/genetics , Nerve Tissue Proteins/genetics , Secretory Vesicles/metabolism , Adult , Aged , Animals , Animals, Genetically Modified , Base Sequence , Blood Platelets/pathology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Male , Middle Aged , Molecular Sequence Data , Nerve Tissue Proteins/antagonists & inhibitors , Pedigree , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Young Adult , Zebrafish/growth & development , Zebrafish/metabolism
13.
Dev Cell ; 20(5): 597-609, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21571218

ABSTRACT

Hematopoietic differentiation critically depends on combinations of transcriptional regulators controlling the development of individual lineages. Here, we report the genome-wide binding sites for the five key hematopoietic transcription factors--GATA1, GATA2, RUNX1, FLI1, and TAL1/SCL--in primary human megakaryocytes. Statistical analysis of the 17,263 regions bound by at least one factor demonstrated that simultaneous binding by all five factors was the most enriched pattern and often occurred near known hematopoietic regulators. Eight genes not previously appreciated to function in hematopoiesis that were bound by all five factors were shown to be essential for thrombocyte and/or erythroid development in zebrafish. Moreover, one of these genes encoding the PDZK1IP1 protein shared transcriptional enhancer elements with the blood stem cell regulator TAL1/SCL. Multifactor ChIP-Seq analysis in primary human cells coupled with a high-throughput in vivo perturbation screen therefore offers a powerful strategy to identify essential regulators of complex mammalian differentiation processes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/metabolism , Genome, Human/genetics , Megakaryocytes/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins/metabolism , Binding Sites , Cell Differentiation , Cells, Cultured , Humans , Megakaryocytes/cytology , Oligonucleotide Array Sequence Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1
14.
Blood ; 115(24): 5069-79, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20351310

ABSTRACT

Exposed subendothelial collagen acts as a substrate for platelet adhesion and thrombus formation after vascular injury. Synthetic collagen-derived triple-helical peptides, designated collagen-related peptide (CRP), GFOGER, and VWF-III, can specifically engage the platelet collagen receptors, glycoprotein VI and integrin alpha(2)beta(1), and plasma von Willebrand factor (VWF), respectively. Hitherto, the role of these 3 collagen-binding axes has been studied indirectly. Use of these uniform peptide substrates, rather than collagen fibers, provides independent control of each axis. Here, we use confocal imaging and novel image analysis techniques to investigate the effects of receptor-ligand engagement on platelet binding and activation during thrombus formation under flow conditions. At low shear (100s(-1) and 300s(-1)), both GFOGER and CRP are required for thrombus formation. At 1000s(-1), a combination of either CRP or GFOGER with VWF-III induces comparable thrombus formation, and VWF-III increases thrombus deposition at all shear rates, being indispensable at 3000s(-1). A combination of CRP and VWF-III is sufficient to support extensive platelet deposition at 3000s(-1), with slight additional effect of GFOGER. Measurement of thrombus height after specific receptor blockade or use of altered proportions of peptides indicates a signaling rather than adhesive role for glycoprotein VI, and primarily adhesive roles for both alpha(2)beta(1) and the VWF axis.


Subject(s)
Blood Platelets/metabolism , Collagen/metabolism , Integrin alpha2beta1/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, Collagen/metabolism , Thrombosis/metabolism , Carrier Proteins/metabolism , Humans , Molecular Mimicry , Peptide Fragments/metabolism , Peptides/metabolism , Platelet Adhesiveness/physiology , Protein Binding/physiology , Regional Blood Flow/physiology , Signal Transduction/physiology , Stress, Mechanical , von Willebrand Factor/metabolism
15.
Blood ; 111(10): 4986-96, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18305222

ABSTRACT

We have analyzed the adhesion of human and murine platelets, and of recombinant human and murine GpVI ectodomains, to synthetic triple-helical collagen-like peptides. These included 57 peptides derived from the sequence of human type III collagen and 9 peptides derived from the cyanogen bromide fragment of bovine type III collagen, alpha1(III)CB4. We have identified several peptides that interact with GpVI, in particular a peptide designated III-30 with the sequence GAOGLRGGAGPOGPEGGKGAAGPOGPO. Both human and murine platelets bound to peptide III-30 in a GpVI-dependent manner. III-30 also supported binding of recombinant GpVI ectodomains. Cross-linked III-30 induced aggregation of human and murine platelets, although with a lower potency than collagen-related peptide. Modifications of the peptide sequence indicated that the hydroxyproline residues play a significant role in supporting its GpVI reactivity. However, many peptides containing OGP/GPO motifs did not support adhesion to GpVI. These data indicate that the ability of a triple-helical peptide to bind GpVI is not solely determined by the presence or spatial arrangement of these OGP/GPO motifs within the peptides.


Subject(s)
Collagen Type III/metabolism , Platelet Membrane Glycoproteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Blood Platelets/metabolism , Cattle , Collagen Type III/chemistry , Humans , Mice , Peptide Fragments , Peptides/chemical synthesis , Peptides/chemistry , Platelet Aggregation , Platelet Membrane Glycoproteins/chemistry , Protein Binding
16.
J Biol Chem ; 282(2): 1296-304, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17085439

ABSTRACT

Collagen-related peptide is a selective agonist for the platelet collagen receptor Glycoprotein VI. The triple helical peptide contains ten GPO triplets/strand (single letter amino acid nomenclature, where O is hydroxyproline) and so over-represents GPO compared with native collagen sequence. To investigate the ability of Glycoprotein VI to recognize GPO triplets in a setting more representative of the collagens, we synthesized a set of triple helical peptides containing fewer GPO triplets, varying their number and spacing within an inert (GPP)n backbone. The adhesion of recombinant human Glycoprotein VI ectodo-main, like that of human platelets, to these peptides increased with their GPO content, and platelet adhesion was abolished by the specific anti-Glycoprotein VI-blocking antibody, 10B12. Platelet aggregation and protein tyrosine phosphorylation were induced only by cross-linked peptides and only those that contained two or more GPO triplets. Such peptides were less potent than cross-linked collagen-related peptide. Our data suggest that both the sequences GPOGPO and GPO.........GPO represent functional Glycoprotein VI recognition motifs within collagen. Furthermore, we propose that the (GPO)4 motif can support simultaneous binding of two glycoprotein VI molecules, in either a parallel or anti-parallel stacking arrangement, which could play an important role in activation of signaling.


Subject(s)
Blood Platelets/metabolism , Collagen/chemistry , Collagen/metabolism , Platelet Adhesiveness/physiology , Platelet Membrane Glycoproteins/metabolism , Binding Sites/physiology , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/metabolism , Glycine/metabolism , Humans , Hydroxyproline/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/chemistry , Proline/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology , Structure-Activity Relationship , Tyrosine/metabolism
17.
J Immunol Methods ; 316(1-2): 75-83, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17027020

ABSTRACT

We report the development of an expression system for the production of soluble, calmodulin (CaM)-tagged proteins in Drosophila Schneider S2 cells and the subsequent use of these proteins for the selection of phage displayed antibodies. The CaM-tag permitted the purification of recombinant protein to >90% purity in a single step at yields of >20 mg/l. Using platelet glycoprotein VI (GP6) as a model, we demonstrated that the recombinant CaM-tagged protein was post-translationally N-glycosylated and had identical ligand specificity to native protein. A novel selection strategy, exploiting the CaM tag, was then used to isolate four single chain Fv fragments (scFvs) specific for GP6 from a non-immune phage display library. In contrast to other selection methods, which can result in antibodies that do not recognise native protein, all of the scFvs we selected bound cell surface expressed GP6. In conclusion, the production of CaM-tagged proteins in Drosophila Schneider S2 cells and the selection strategy reported here offer advantages over previously published methods, including simple culture conditions, rapid protein purification, specific elution of phage antibodies and preferential selection of phage antibodies that recognise native, cell surface expressed protein.


Subject(s)
Antigens/biosynthesis , Calmodulin/genetics , Drosophila melanogaster/genetics , Immunoglobulin Fragments/biosynthesis , Peptide Library , Platelet Membrane Glycoproteins/genetics , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Antigens/genetics , Blotting, Western , Calmodulin/metabolism , Cell Line , Cloning, Molecular , Drosophila melanogaster/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Molecular Sequence Data , Platelet Membrane Glycoproteins/immunology , Recombinant Proteins/chemical synthesis , Recombinant Proteins/genetics
18.
J Biol Chem ; 281(44): 33505-10, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16956881

ABSTRACT

Platelet activation by collagen relies on the interaction of the receptor glycoprotein VI (GPVI) with collagen helices. We have previously generated two recombinant single chain human antibodies (scFvs) to human GPVI. The first, 10B12, binds to the collagen-binding site on the apical surface between the two immunoglobulin-like domains (D1D2) of the receptor and so directly inhibits GPVI function. The second, 1C3, binds D1D2 independently of 10B12 and has been shown to have a more subtle effect on platelet responses to collagen. Here we have shown that 1C3 potentiates the effect of 10B12 on platelet aggregation induced by collagen and cross-linked collagen-related peptide (CRP-XL). We investigated this by measuring the effect of both scFvs on the binding of D1D2 to immobilized collagen and CRP. As expected, 10B12 completely inhibited binding of GPVI to each ligand in a dose-dependent manner. However, 1C3 inhibited only a proportion of GPVI binding to its ligands, implying that it interferes with another aspect of ligand recognition by GPVI. To further understand the mode of inhibition, we used a unique set of CRPs in which the content of critical glycine-proline-hydroxyproline (GPO) triplets was varied in relation to an "inert" scaffold sequence of GPP motifs. We observed that a stepwise increase in D1D2 binding with (GPO)(2) content was blocked by 1C3. Together these results indicate that 1C3 inhibits clustering of the immunoglobulin-like domains of GPVI on collagen/CRPs, a conclusion that is supported by mapping the 1C3 epitope to the region including isoleucine 148 in D2.


Subject(s)
Collagen/metabolism , Platelet Membrane Glycoproteins/metabolism , Antibodies/immunology , Epitopes/immunology , Humans , Isoleucine/genetics , Isoleucine/metabolism , Models, Molecular , Mutation/genetics , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Platelet Aggregation , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Protein Binding , Protein Structure, Tertiary
19.
FASEB J ; 19(8): 898-909, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923400

ABSTRACT

Lipid-rich atherosclerotic plaques are vulnerable, and their rupture can cause the formation of a platelet- and fibrin-rich thrombus leading to myocardial infarction and ischemic stroke. Although the role of plaque-based tissue factor as stimulator of blood coagulation has been recognized, it is not known whether plaques can cause thrombus formation through direct activation of platelets. We isolated lipid-rich atheromatous plaques from 60 patients with carotid stenosis and identified morphologically diverse collagen type I- and type III-positive structures in the plaques that directly stimulated adhesion, dense granule secretion, and aggregation of platelets in buffer, plasma, and blood. This material also elicited platelet-monocyte aggregation and platelet-dependent blood coagulation. Plaques exposed to flowing blood at arterial wall shear rate induced platelets to adhere to and spread on the collagenous structures, triggering subsequent thrombus formation. Plaque-induced platelet thrombus formation was observed in fully anticoagulated blood (i.e., in the absence of tissue factor-mediated coagulation). Mice platelets lacking glycoprotein VI (GPVI) were unable to adhere to atheromatous plaque or form thrombi. Human platelet thrombus formation onto plaques in flowing blood was completely blocked by GPVI inhibition with the antibody 10B12 but not affected by integrin alpha2beta1 inhibition with 6F1 mAb. Moreover, the initial platelet response, shape change, induced by plaque was blocked by GPVI inhibition but not with alpha2beta1 antagonists (6F1 mAb or GFOGER-GPP peptide). Pretreatment of plaques with collagenase or anti-collagen type I and anti-collagen type III antibodies abolished plaque-induced platelet activation. Our results indicate that morphologically diverse collagen type I- and collagen type III-containing structures in lipid-rich atherosclerotic plaques stimulate thrombus formation by activating platelet GPVI. This platelet collagen receptor, essential for plaque-induced thrombus formation, presents a promising new anti-thrombotic target for the prevention of ischemic cardiovascular diseases.


Subject(s)
Atherosclerosis/complications , Platelet Membrane Glycoproteins/physiology , Thrombosis/etiology , Animals , Atherosclerosis/pathology , Blood Coagulation , Blood Platelets/physiology , Carotid Stenosis/blood , Collagen Type I/analysis , Collagen Type III/analysis , Humans , Integrin alpha2beta1/physiology , Lipids/analysis , Mice , Microscopy, Fluorescence , Monocytes/physiology , Platelet Activation , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/antagonists & inhibitors
20.
J Biol Chem ; 279(46): 47763-72, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15345717

ABSTRACT

Only three recognition motifs, GFOGER, GLOGER, and GASGER, all present in type I collagen, have been identified to date for collagen-binding integrins, such as alpha(2)beta(1). Sequence alignment was used to investigate the occurrence of related motifs in other human fibrillar collagens, and located a conserved array of novel GER motifs within their triple helical domains. We compared the integrin binding properties of synthetic triple helical peptides containing examples of such sequences (GLSGER, GMOGER, GAOGER, and GQRGER) or the previously identified motifs. Recombinant inserted (I) domains of integrin subunits alpha(1), alpha(2) and alpha(11) all bound poorly to all motifs other than GFOGER and GLOGER. Similarly, alpha(2)beta(1) -containing resting platelets adhered well only to GFOGER and GLOGER, while ADP-activated platelets, HT1080 cells and two active alpha(2)I domain mutants (E318W, locked open) bound all motifs well, indicating that affinity modulation determines the sequence selectivity of integrins. GxO/SGER peptides inhibited platelet adhesion to collagen monomers with order of potency F >/= L >/= M > A. These results establish GFOGER as a high affinity sequence, which can interact with the alpha(2)I domain in the absence of activation and suggest that integrin reactivity of collagens may be predicted from their GER content.


Subject(s)
Amino Acid Motifs , Collagen Type I/metabolism , Integrin alpha2beta1/metabolism , Peptides/metabolism , Animals , Blood Platelets/metabolism , Cell Line , Collagen Type I/chemistry , Collagen Type I/genetics , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Integrin alpha2beta1/genetics , Peptides/chemistry , Peptides/genetics , Platelet Adhesiveness/physiology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...