Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter Horsewood is active.

Publication


Featured researches published by Peter Horsewood.


The New England Journal of Medicine | 1995

Heparin-Induced Thrombocytopenia in Patients Treated with Low-Molecular-Weight Heparin or Unfractionated Heparin

Theodore E. Warkentin; Mark N. Levine; Jack Hirsh; Peter Horsewood; Robin S. Roberts; Michael Gent; John G. Kelton

BACKGROUND Heparin-induced thrombocytopenia, defined by the presence of heparin-dependent IgG antibodies, typically occurs five or more days after the start of heparin therapy and can be complicated by thrombotic events. The frequency of heparin-induced thrombocytopenia and of heparin-dependent IgG antibodies, as well as the relative risk of each in patients given low-molecular-weight heparin, is unknown. METHODS We obtained daily platelet counts in 665 patients in a randomized, double-blind clinical trial comparing unfractionated heparin with low-molecular-weight heparin as prophylaxis after hip surgery. Heparin-induced thrombocytopenia was defined as a decrease in the platelet count below 150,000 per cubic millimeter that began five or more days after the start of heparin therapy, and a positive test for heparin-dependent IgG antibodies. We also tested a representative subgroup of 387 patients for heparin-dependent IgG antibodies regardless of their platelet counts. RESULTS Heparin-induced thrombocytopenia occurred in 9 of 332 patients who received unfractionated heparin and in none of 333 patients who received low-molecular-weight heparin (2.7 percent vs. 0 percent; P = 0.0018). Eight of the 9 patients with heparin-induced thrombocytopenia also had one or more thrombotic events (venous in 7 and arterial in 1), as compared with 117 of 656 patients without heparin-induced thrombocytopenia (88.9 percent vs. 17.8 percent; odds ratio, 36.9; 95 percent confidence interval, 4.8 to 1638; P < 0.001). In the subgroup of 387 patients, the frequency of heparin-dependent IgG antibodies was higher among patients who received unfractionated heparin (7.8 percent, vs. 2.2 percent among patients who received low-molecular-weight heparin; P = 0.02). CONCLUSIONS Heparin-induced thrombocytopenia, associated thrombotic events, and heparin-dependent IgG antibodies are more common in patients treated with unfractionated heparin than in those treated with low-molecular-weight heparin.


Journal of Clinical Investigation | 1993

Multimerin is found in the alpha-granules of resting platelets and is synthesized by a megakaryocytic cell line.

C. P. M. Hayward; Dorothy F. Bainton; James W. Smith; Peter Horsewood; R. H. Stead; T. J. Podor; Theodore E. Warkentin; John G. Kelton

In this report, we describe the intracellular localization of multimerin in platelets and its biosynthesis by Dami cells, a megakaryocytic cell line. Immunoelectron microscopy was used to examine frozen thin sections of resting and activated platelets. Multimerin was localized within the platelet alpha-granule in an eccentric position. Within activated platelets, multimerin was found in the surface-connected open cannalicular system and on the external plasma membrane. Light microscopic immunocytochemistry demonstrated multimerin in normal megakaryocytes and in Dami cells after stimulation with PMA. Confirmation of multimerin biosynthesis by Dami cells was obtained by metabolic labeling studies. Both platelet and Dami cell multimerin demonstrated several subunit sizes on reduced SDS-PAGE. However, peptide mapping confirmed structural homology between the different multimerin subunits. Glycosidase digestion demonstrated that multimerin is heavily glycosylated with mainly complex, N-linked carbohydrate. In contrast to the multimerin isolated from platelets, cultured Dami cells secreted mainly smaller multimers of the protein. Biosynthesis of multimerin by a megakaryocytic cell line supports endogenous biosynthesis by megakaryocytes as the origin of this platelet alpha-granule protein.


Journal of Laboratory and Clinical Medicine | 1998

ABH antigens on human platelets : expression on the glycosyl phosphatidylinositol-anchored protein CD109

John G. Kelton; James W. Smith; Peter Horsewood; Margaret N. Warner; Theodore E. Warkentin; Robert W. Finberg; Catherine P. M. Hayward

Platelets express alloantigens that are platelet specific (eg, the HPA antigens) and alloantigens that are shared with other blood cells (eg, the ABH antigens). The blood group A and B determinants are expressed on glycolipids and on some intrinsic platelet membrane glycoproteins. This report characterizes multiple platelet proteins reacting with blood group antibodies in serum samples from mothers of children born with neonatal alloimmune thrombocytopenia. ABH antigens on additional platelet proteins are identified, including the glycosyl phosphatidylinositol-anchored protein CD109. The proteins that carry ABH antigens were identified by using monoclonal antibodies to glycoproteins Ib, IIb/IIIa, Ia/IIa, CD31, and CD109 and immunoprecipitation/immunoblotting techniques with monoclonal antibodies to A and B antigens. The maternal serum samples and anti-A and anti-B monoclonal antibodies immunoprecipitated identical radiolabeled platelet proteins including proteins at 220 and 175 kd and proteins with mobilities corresponding to glycoproteins Ib, IIb/IIIa, IV, and V. Treatment of platelets with phosphatidylinositol-specific phospholipase C released into the supernatant a 175-kd protein that expressed the blood group determinants. This protein comigrated with the glycosyl phosphatidylinositol-anchored protein CD109. When platelet proteins were purified by immunoprecipitation with monoclonal antibodies and then tested by immunoblotting, anti-A reacted with the glycosyl phosphatidylinositol-anchored protein CD109 and to glycoproteins Ib, IIb, IIa, IIIa, and CD31 (PECAM). These results indicate that structures for modification by glycosyltransferases exist on platelet CD109, which also expresses the Gov alloantigen system. This study indicates that certain platelet proteins express both platelet-specific and blood group antigens that may contribute to platelet transfusion refractoriness and to neonatal alloimmune thrombocytopenia.


Transfusion | 1984

Quantitation of red cell-associated IgG using an immunoradiometric assay

M. O. Jeje; M. A. Blajchman; K. Steeves; Peter Horsewood; John G. Kelton

In this report, we describe a sensitive immunoradiometric assay (IRMA) for quantitating IgG on the surface of red cells. Washed red cells were prepared to a purity of greater than 99.9 percent. Varying dilutions of these cells were incubated with a fixed concentration of 125I‐anti‐IgG. After equilibrium was achieved, the unbound 125I‐anti‐IgG was measured by the addition of IgG covalently linked to agarose beads. The red cells were lysed by detergent, and the 125I‐anti‐IgG bound to the IgG‐ beads was measured. The amount of IgG on the red cells was determined by relating the concentration of test red cells causing 50 percent inhibition of binding of the 125I‐anti‐IgG to the IgG‐beads to 50 percent inhibition of binding caused by the IgG standard. Using this assay, the red cell‐associated IgG (RCA‐IgG) of 20 healthy male and female controls with normal hemoglobin concentrations was 7.23 +/− 6.11 fg IgG per 10(3) cells (mean +/− 2 SD). The mean RCA‐IgG on washed cells from 34 different tests performed on 19 anemic patients with clinically diagnosed autoimmune hemolytic anemia was 176.1 +/− 375.6 fg IgG per 10(3) cells. There was no correlation between the levels of RCA‐ IgG and the hemoglobin levels or reticulocyte counts in these patients.


British Journal of Haematology | 1989

Platelet specific alloantigens on the platelet glycoprotein Ia/IIa complex

James W. Smith; John G. Kelton; Peter Horsewood; Claudia Brown; Alan R. Giles; Ralph M. Meyer; Virgil L. Woods; Robert F. Burrows

Summary The majority of platelet alloantigens are located on platelet glycoproteins IIb/IIIa. This report describes a codominant allelic system carried on the glycoprotein la/IIa complex, which we originally designated as Zava/Zavb but which is identical to the Bra/Brb system. Furthermore Zava was found to be identical to Hca. The alloantigens could not be detected using a direct binding enzyme immunoassay (EIA) with intact platelets, but were readily detected using a glycoprotein capture EIA and by radioimmunoprecipitation techniques. The two index cases (designated as homozygous Zava and Zavb) had alloantibodies against the corresponding antigen and did not react with their own platelets. Using these alloantibodies and a monoclonal antibody that reacts with the platelet glycoprotein la/IIa complex (12F1). We demonstrated that all Ia/IIa molecules carry either Zava or Zavb and we found that Zava and Zavb are on discrete populations of Ia/IIa. Following immunodepletion using either anti‐Zava or anti‐Zavb, all detectable Ia/IIa complexes from the respective homozygous platelets were removed. Immunodepletion of heterozygous Zava/Zavb with either anti‐Zava or anti‐Zavb did not reduce the amount of Ia/IIa complexes precipitable using the alternate alloantiserum. Population studies (n= 50) indicated the phenotypic frequency of Zava/Zava is less than 1%; Zava/Zavb is 18% and Zavb/Zavb is 82%. Four different alloantisera that had either anti‐Zava or anti‐Zavb reactivity also carried reactivity against the Baka or Bakb antigens which may suggest an association in the immune response to these alleles.


Journal of Immunological Methods | 1993

Investigation of human platelet alloantigens and glycoproteins using non-radioactive immunoprecipitation.

James W. Smith; C. P. M. Hayward; Theodore E. Warkentin; Peter Horsewood; John G. Kelton

Sensitive techniques to detect platelet antibodies are needed for the investigation of immune thrombocytopenic syndromes such as neonatal alloimmune thrombocytopenia and post-transfusion purpura. Radioimmunoprecipitation has proved useful in the investigation of platelet-antibody interactions; however, the requirement for a radioactive label is a disadvantage. We describe the immunoprecipitation of human platelet proteins labelled with nonradioactive NHSS-biotin and compare the results with proteins labelled with 125I. The efficiency of labelling was evaluated by immunoprecipitation using well-characterized human anti-platelet antisera and murine monoclonal antibodies. The immunoprecipitated proteins were separated by SDS-PAGE, transferred to nitrocellulose and detected using streptavidin-horseradish peroxidase and a chemiluminescent substrate with exposure to X ray film. The biotinylation technique labelled glycoproteins Ia/IIa, Ib/IX, IIb/IIIa, IV, and p175 which carry all of the known platelet alloantigens and isoantigens. It was as sensitive as radiolabelling and had the advantage of labelling GPs Ib beta and IX, which were not labelled using radioiodine. Human sera containing alloantibodies to HPA-1a on GP IIIa, HPA-3a on GP IIb, HPA-5a and HPA-5b on GP Ia, Govb on p175, and the isoantibody Naka on GP IV precipitated the corresponding biotinylated proteins. Biotinylated proteins could be detected using a 30 s exposure compared to 2 days or longer for 125I. Immunoprecipitation of human platelet glycoproteins labeled with NHSS-biotin is a fast and sensitive alternative to conventional radioimmunoprecipitation for the study of human platelet antigens.


British Journal of Haematology | 1995

The prenatal identification of fetal compatibility in neonatal alloimmune thrombocytopenia using amniotic fluid and variable number of tandem repeat (VNTR) analysis

Gregory A. Denomme; John S. Waye; Robert F. Borrows; Catherine P. M. Hayward; Theodore E. Warkentin; Peter Horsewood; James W. Smith; Russel D. Jelsema; Laura J. Zuidema; John G. Kelton

Summary. Most severe episodes of neonatal alloimmune thrombocytopenic purpura (NATP) are caused by antiplatelet alloantibodies against the HPA‐la (PlA1) antigen. However, half of subsequent fetuses produced from a HPA‐la/b father (genotypic frequency 28%) will result in a child who is not affected. Some investigators manage NATP by confirming the fetal platelet phenotype using percutaneous umbilical cord sampling, a procedure that carries a low but real risk of fetal morbidity and mortality. More recently, physicians determine the fetal platelet antigen genotype using DNA derived from amniotic fluid or chorionic villus samples. All therapy is withdrawn for a fetus who genotypes as HPA‐lb/b. However, since the fetus is the same genotype as the mother, there can be uncertainty about the origin of the genetic material and thus the validity of the fetal genotype. The inappropriate withdrawal of therapy for a erroneously genotyped fetus could be fatal, and consequently many physicians advocate fetal HPA‐1 phenotyping with confirmation using percutaneous umbilical blood sampling.


Journal of Immunoassay | 1983

The Measurement of Plateiet-Associated IgG Using an Immunoradiometric Assay

John G. Kelton; Greg Denorame; Chris Walker; Peter Horsewood; Jack Gauldie

The amount of IgG on the surface of washed platelets from healthy individuals varies according to the assay used. An immunoradiometric assay (IRMA) for IgG was developed, validated and used to measure platelet associated IgG. The platelet-associated IgG (PAIgG) on washed platelets was allowed to react with excess 125I-labelled anti-IgG. The unbound 125I-anti-IgG was then quantitated by the addition of sepharose beads to which IgG had been covalently bound. The 125I-anti-IgG/IgG-beads were separated from the platelet suspension by passage across a density gradient. The mean amount of IgG present on washed platelets from healthy individuals was 0.8 fg IgG/platelet, or approximately 4,000 molecules of IgG per cell. Inverse Scatchard analysis confirmed the validity of the assay calibration and identical results were obtained when five different anti-IgG antibodies were used. The specific binding of 125I-anti-IgG to platelet-bound IgG was complete by 30 min, but non-specific binding of radioactivity continued thereafter. This non-specific binding occurred not only with anti-IgG but with all antibodies tested and could give elevated estimates of PAIgG in direct binding assays.


Journal of Thrombosis and Haemostasis | 2013

FcγRIIa proteolysis as a diagnostic biomarker for heparin‐induced thrombocytopenia

Ishac Nazi; Donald M. Arnold; James W. Smith; Peter Horsewood; Jane C. Moore; Theodore E. Warkentin; Mark Crowther; John G. Kelton

A significant challenge in the management of heparin‐induced thrombocytopenia (HIT) patients is making a timely and accurate diagnosis. The readily available enzyme immunoassays (EIAs) have low specificities. In contrast, platelet activation assays have higher specificities, but they are technically demanding and not widely available. In addition, ~ 10% of samples referred for HIT testing are initially classified as indeterminate by the serotonin release assay (SRA), which further delays accurate diagnosis. HIT is characterized by platelet activation, which leads to FcγRIIa proteolysis. This raises the possibility that identification of the proteolytic fragment of FcγRIIa could serve as a surrogate marker for HIT.


Platelets | 2000

Investigation of a platelet factor 4 polymorphism on the immune response in patients with heparin-induced thrombocytopenia

Peter Horsewood; John G. Kelton

A small fraction of patients who receive heparin develop heparin-induced thrombocytopenia (HIT) and, of these patients, a still smaller proportion develop associated thrombotic complications. Heparin-induced thrombocytopenia is caused by the formation of antibodies that bind to specific complexes of platelet factor 4 (PF4) and heparin. However, it remains uncertain why certain patients form these antibodies and develop HIT or why certain patients have thrombotic events. In this report we describe studies on individuals with and without HIT to determined if a potential PF4 polymorphism could explain differences in susceptibility to HIT. In the 10 control individuals and the 10 patients we studied, we did not find a difference in the PF4 sequences. Genetic difference in the PF4 antigenic target does not explain the occurrence of HIT in susceptible patients.

Collaboration


Dive into the Peter Horsewood's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. P. M. Hayward

McMaster University Medical Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory A. Denomme

Gulf Coast Regional Blood Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge