Network


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

Hotspot


Dive into the research topics where R. McMaster is active.

Publication


Featured researches published by R. McMaster.


Science Signaling | 2009

Leishmania GP63 Alters Host Signaling Through Cleavage-Activated Protein Tyrosine Phosphatases

Maria Adelaida Gomez; Irazú Contreras; Maxime Hallé; Michel L. Tremblay; R. McMaster; Martin Olivier

The parasite protein GP63 triggers cleavage and activation of host protein tyrosine phosphatases to promote infection. Exploiting the Host’s Phosphatases Leishmaniasis is a globally important infectious disease caused by the parasite Leishmania. Gomez et al. show that infection of macrophages with Leishmania alters the activity of multiple protein tyrosine phosphatases (PTPs) through cleavage mediated by the parasite protein GP63. The activated PTPs inhibit macrophage inflammatory immune responses through dephosphorylation of Janus kinases. In addition to the PTP SHP-1, previously reported to be activated in response to Leishmania infection, Gomez et al. show that the PTPs TCPTP and PTP1B are also activated and that PTP1B serves a key role in the initial stages of disease progression in mice. With more than 12 million people affected worldwide, 2 million new cases occurring per year, and the rapid emergence of drug resistance and treatment failure, leishmaniasis is an infectious disease for which research on drug and vaccine development, host-pathogen, and vector-parasite interactions are current international priorities. Upon Leishmania-macrophage interaction, activation of the protein tyrosine phosphatase (PTP) SHP-1 rapidly leads to the down-regulation of Janus kinase and mitogen-activated protein kinase signaling, resulting in the attenuation of host innate inflammatory responses and of various microbicidal macrophage functions. We report that, in addition to SHP-1, the PTPs PTP1B and TCPTP are activated and posttranslationally modified in infected macrophages, and we identify an essential role for PTP1B in the in vivo progression of Leishmania infection. The mechanism underlying PTP modulation involves the proteolytic activity of the Leishmania surface protease GP63. Access of GP63 to macrophage PTP1B, TCPTP, and SHP-1 is mediated in part by a lipid raft–dependent mechanism, resulting in PTP cleavage and stimulation of phosphatase activity. Collectively, our data present a mechanism of cleavage-dependent activation of macrophage PTPs by an obligate intracellular pathogen and show that internalization of GP63, a key Leishmania virulence factor, into host macrophages is a strategy the parasite uses to interact and survive within its host.


Cell Host & Microbe | 2011

Leishmania Repression of Host Translation through mTOR Cleavage Is Required for Parasite Survival and Infection

Maritza Jaramillo; Maria Adelaida Gomez; Ola Larsson; Marina Tiemi Shio; Ivan Topisirovic; Irazú Contreras; Randi Luxenburg; Amy B. Rosenfeld; Rodney Colina; R. McMaster; Martin Olivier; Mauro Costa-Mattioli; Nahum Sonenberg

The protozoan parasite Leishmania alters the activity of its host cell, the macrophage. However, little is known about the effect of Leishmania infection on host protein synthesis. Here, we show that the Leishmania protease GP63 cleaves the mammalian/mechanistic target of rapamycin (mTOR), a serine/threonine kinase that regulates the translational repressor 4E-BP1. mTOR cleavage results in the inhibition of mTOR complex 1 (mTORC1) and concomitant activation of 4E-BP1 to promote Leishmania proliferation. Consistent with these results, pharmacological activation of 4E-BPs with rapamycin, results in a dramatic increase in parasite replication. In contrast, genetic deletion of 4E-BP1/2 reduces parasite load in macrophages ex vivo and decreases susceptibility to cutaneous leishmaniasis in vivo. The parasite resistant phenotype of 4E-BP1/2 double-knockout mice involves an enhanced type I IFN response. This study demonstrates that Leishmania evolved a survival mechanism by activating 4E-BPs, which serve as major targets for host translational control.


PLOS Pathogens | 2010

Leishmania-Induced Inactivation of the Macrophage Transcription Factor AP-1 Is Mediated by the Parasite Metalloprotease GP63

Irazú Contreras; Maria Adelaida Gomez; Oliver Nguyen; Marina Tiemi Shio; R. McMaster; Martin Olivier

Leishmania parasites have evolved sophisticated mechanisms to subvert macrophage immune responses by altering the host cell signal transduction machinery, including inhibition of JAK/STAT signalling and other transcription factors such as AP-1, CREB and NF-κB. AP-1 regulates pro-inflammatory cytokines, chemokines and nitric oxide production. Herein we show that upon Leishmania infection, AP-1 activity within host cells is abolished and correlates with lower expression of 5 of the 7 AP-1 subunits. Of interest, c-Jun, the central component of AP-1, is cleaved by Leishmania. Furthermore, the cleavage of c-Jun is dependent on the expression and activity of the major Leishmania surface protease GP63. Immunoprecipitation of c-Jun from nuclear extracts showed that GP63 interacts, and cleaves c-Jun at the perinuclear area shortly after infection. Phagocytosis inhibition by cytochalasin D did not block c-Jun down-regulation, suggesting that internalization of the parasite might not be necessary to deliver GP63 molecules inside the host cell. This observation was corroborated by the maintenance of c-Jun cleavage upon incubation with L. mexicana culture supernatant, suggesting that secreted, soluble GP63 could use a phagocytosis-independent mechanism to enter the host cell. In support of this, disruption of macrophage lipid raft microdomains by Methyl β-Cyclodextrin (MβCD) partially inhibits the degradation of full length c-Jun. Together our results indicate a novel role of the surface protease GP63 in the Leishmania-mediated subversion of host AP-1 activity.


Experimental Parasitology | 1990

Leishmania major: Production of recombinant gp63, its antigenicity and immunogenicity in mice

Emanuela Handman; Linda L. Button; R. McMaster

The Mr 63,000 membrane polypeptide (gp63) is one of the Leishmania receptors for host macrophages and has been shown to protect mice from infection. The gene encoding gp63, the major Mr 63,000 surface glycoprotein of L. major promastigotes, has been expressed as a fusion protein with the enzyme glutathione S- transferase encoded by the parasitic helminth Schistosoma japonicum. This fusion protein was recognized by polyclonal antibodies to the native Leishmania gp63 polypeptide. The insoluble gp63 fusion protein was purified by SDS-PAGE and electroelution and was used to raise antibodies in rabbits. These rabbit anti-gp63 antibodies recognized the fusion protein and the denatured parasite gp63 on immunoblots and by immunofluorescence on fixed promastigotes, but did not recognize the native molecule on live organisms. However, antibodies raised against native promastigote glycoproteins, affinity purified on solid-phase gp63 fusion protein, recognized both native and denatured gp63, suggesting the presence of native determinants in the recombinant protein. The gp63 fusion protein did not protect mice of either healer or nonhealer phenotype from challenge infection with live promatigotes. The implications of these results for the engineering of recombinant DNA-produced molecular vaccines are discussed.


Journal of Heart and Lung Transplantation | 2009

Whole Blood Genomic Biomarkers of Acute Cardiac Allograft Rejection

David Lin; Zsuzsanna Hollander; Raymond T. Ng; C. Imai; Andrew Ignaszewski; Robert Balshaw; Gabriela V. Cohen Freue; J. Wilson-McManus; Pooran Qasimi; Anna Meredith; Alice Mui; Timothy J. Triche; R. McMaster; Paul Keown; Bruce M. McManus

BACKGROUND Significant progress has been made in cardiac transplantation over the past 30 years; however, the means for detection of acute cardiac allograft rejection remains in need of improvement. At present, the endomyocardial biopsy, an invasive and inconvenient procedure for patients, is required for the surveillance and diagnosis of acute cardiac allograft rejection. In the Biomarkers in Transplantation initiative, we investigated gene expression profiles in peripheral blood of cardiac transplant subjects as potential biomarkers for diagnosis of allograft rejection. METHODS Whole blood samples were obtained from 28 cardiac transplant subjects who consented to the study. Serial samples were collected from pre-transplant through 3 years post-transplant according to the standard protocol. Temporally correspondent biopsies were also collected, reviewed in a blinded manner, and graded according to current ISHLT guidelines. Blood samples were analyzed using Affymetrix microarrays. Genomic profiles were compared in subjects with acute rejection (AR; ISHLT Grade > or =2R) and no rejection (NR; Grade 0R). Biomarker panel genes were identified using linear discriminant analysis. RESULTS We found 1,295 differentially expressed probe-sets between AR and NR samples and developed a 12-gene biomarker panel that classifies our internal validation samples with 83% sensitivity and 100% specificity. CONCLUSIONS Based on our current results, we believe whole blood genomic biomarkers hold great potential in the diagnosis of acute cardiac allograft rejection. A prospective, Canada-wide trial will be conducted shortly to further evaluate the classifier panel in diverse patients and a range of clinical programs.


Vaccine | 1996

Effective immunization of mice against cutaneous leishmaniasis using an intrinsically adjuvanted synthetic lipopeptide vaccine

Shoshana Frankenburg; Ofra Axelrod; Shirley Kutner; Charles L. Greenblatt; Sidney Klaus; Esther Aboud Pirak; R. McMaster; George H. Lowell

Two peptides representing predicted T-cell epitopes of gp63, a major surface glycoprotein of the parasite Leishmania major, were used in vaccines tested in a murine model of cutaneous leishmaniasis. Either subcutaneous or intraperitoneal immunization in saline with a peptide representing gp63 amino acids 467-482 (p467) significantly protected CBA mice against the development of severe cutaneous lesions only when the peptide was intrinsically adjuvanted by covalently adding a lauryl-cysteine moiety (LC-p467) to its amino terminus during synthesis. In marked contrast, administration of p467 alone, cysteinyl-p467 or gp63 protein in saline resulted in some disease exacerbation. Splenic cells of LC-p467 immunized mice stimulated in vitro with LC-p467 displayed strong proliferative responses and secretion of IL-2, IFN-tau and GM-CSF (but not IL-4 and IL-10) suggesting that immunization with the lipopeptide induced the TH1 type cytokine responses associated with cell-mediated immunity. The safety, efficacy, ease of production and standardization of such lipopeptide vaccines suggest that they have significant potential for the development of vaccines for humans against leishmaniasis or other parasitic or viral diseases that require cell-mediated immunity for protection.


Transplantation | 2010

Whole blood biomarkers of acute cardiac allograft rejection: double-crossing the biopsy.

Zsuzsanna Hollander; David Lin; Virginia Chen; Raymond T. Ng; J. Wilson-McManus; Andrew Ignaszewski; Gabriela V. Cohen Freue; Rob Balshaw; Alice Mui; R. McMaster; Paul Keown; Bruce M. McManus

Background. Acute rejection is still a significant barrier to long-term survival of the allograft. Current acute rejection diagnostic methods are not specific enough or are invasive. There have been a number of studies that have explored the blood or the biopsy to discover genomic biomarkers of acute rejection; however, none of the studies to date have used both. Methods. We analyzed endomyocardial biopsy tissue and whole blood-derived messenger RNA from 11 acute rejection and 20 nonrejection patients using Affymetrix Human Genome U133 Plus 2.0 chips. We used a novel approach and gained insight into the biology of rejection based on gene expression in the biopsy, and applied this knowledge to the blood analysis to identify novel blood biomarkers. Results. We identified probesets that are differentially expressed between acute rejection and nonrejection patients in the biopsy and blood, and developed three biomarker panels: (1) based on biopsy-only (area under the curve=0.85), (2) based on biopsy-targeted whole blood (area under the curve=0.83), and (3) based on whole blood-only (area under the curve=0.60) analyses. Conclusions. Most of the probesets replicated between biopsy and blood are regulated in opposite direction between the two sources of information. We also observed that the biopsy-targeted blood biomarker discovery approach can improve performance of the biomarker panel. The biomarker panel developed using this targeted approach is able to diagnose acute cardiac allograft rejection almost as well as the biopsy-only based biomarker panel.


Journal of Heart and Lung Transplantation | 2013

Predicting acute cardiac rejection from donor heart and pre-transplant recipient blood gene expression

Zsuzsanna Hollander; Virginia Chen; Keerat Sidhu; David Lin; Raymond T. Ng; Robert Balshaw; Gabriela Cohen-Freue; Andrew Ignaszewski; C. Imai; A. Kaan; Scott J. Tebbutt; J. Wilson-McManus; R. McMaster; Paul Keown; Bruce M. McManus

BACKGROUND Acute rejection in cardiac transplant patients remains a contributory factor to limited survival of implanted hearts. Currently, there are no biomarkers in clinical use that can predict, at the time of transplantation, the likelihood of post-transplant acute cellular rejection. Such a development would be of great value in personalizing immunosuppressive treatment. METHODS Recipient age, donor age, cold ischemic time, warm ischemic time, panel-reactive antibody, gender mismatch, blood type mismatch and human leukocyte antigens (HLA-A, -B and -DR) mismatch between recipients and donors were tested in 53 heart transplant patients for their power to predict post-transplant acute cellular rejection. Donor transplant biopsy and recipient pre-transplant blood were also examined for the presence of genomic biomarkers in 7 rejection and 11 non-rejection patients, using non-targeted data mining techniques. RESULTS The biomarker based on the 8 clinical variables had an area under the receiver operating characteristic curve (AUC) of 0.53. The pre-transplant recipient blood gene-based panel did not yield better performance, but the donor heart tissue gene-based panel had an AUC = 0.78. A combination of 25 probe sets from the transplant donor biopsy and 18 probe sets from the pre-transplant recipient whole blood had an AUC = 0.90. Biologic pathways implicated include VEGF- and EGFR-signaling, and MAPK. CONCLUSIONS Based on this study, the best predictive biomarker panel contains genes from recipient whole blood and donor myocardial tissue. This panel provides clinically relevant prediction power and, if validated, may personalize immunosuppressive treatment and rejection monitoring.


PLOS ONE | 2014

Two-Stage, In Silico Deconvolution of the Lymphocyte Compartment of the Peripheral Whole Blood Transcriptome in the Context of Acute Kidney Allograft Rejection

Casey P. Shannon; Robert Balshaw; Raymond T. Ng; J. Wilson-McManus; Paul Keown; R. McMaster; Bruce M. McManus; David Landsberg; Nicole M. Isbel; Greg Knoll; Scott J. Tebbutt

Acute rejection is a major complication of solid organ transplantation that prevents the long-term assimilation of the allograft. Various populations of lymphocytes are principal mediators of this process, infiltrating graft tissues and driving cell-mediated cytotoxicity. Understanding the lymphocyte-specific biology associated with rejection is therefore critical. Measuring genome-wide changes in transcript abundance in peripheral whole blood cells can deliver a comprehensive view of the status of the immune system. The heterogeneous nature of the tissue significantly affects the sensitivity and interpretability of traditional analyses, however. Experimental separation of cell types is an obvious solution, but is often impractical and, more worrying, may affect expression, leading to spurious results. Statistical deconvolution of the cell type-specific signal is an attractive alternative, but existing approaches still present some challenges, particularly in a clinical research setting. Obtaining time-matched sample composition to biologically interesting, phenotypically homogeneous cell sub-populations is costly and adds significant complexity to study design. We used a two-stage, in silico deconvolution approach that first predicts sample composition to biologically meaningful and homogeneous leukocyte sub-populations, and then performs cell type-specific differential expression analysis in these same sub-populations, from peripheral whole blood expression data. We applied this approach to a peripheral whole blood expression study of kidney allograft rejection. The patterns of differential composition uncovered are consistent with previous studies carried out using flow cytometry and provide a relevant biological context when interpreting cell type-specific differential expression results. We identified cell type-specific differential expression in a variety of leukocyte sub-populations at the time of rejection. The tissue-specificity of these differentially expressed probe-set lists is consistent with the originating tissue and their functional enrichment consistent with allograft rejection. Finally, we demonstrate that the strategy described here can be used to derive useful hypotheses by validating a cell type-specific ratio in an independent cohort using the nanoString nCounter assay.


European Journal of Heart Failure | 2014

Proteomic biomarkers of recovered heart function

Zsuzsanna Hollander; Marie Lazárová; Karen K.Y. Lam; Andrew Ignaszewski; Gavin Y. Oudit; Jason R. B. Dyck; George E. Schreiner; Julie Pauwels; Virginia Chen; Gabriela V. Cohen Freue; Raymond T. Ng; J. Wilson-McManus; Robert Balshaw; Scott J. Tebbutt; R. McMaster; Paul Keown; Bruce M. McManus

Chronic heart failure is a costly epidemic that affects up to 2% of people in developed countries. The purpose of this study was to discover novel blood proteomic biomarker signatures of recovered heart function that could lead to more effective heart failure patient management by both primary care and specialty physicians.

Collaboration


Dive into the R. McMaster's collaboration.

Top Co-Authors

Avatar

Paul Keown

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Raymond T. Ng

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Bruce M. McManus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert Balshaw

BC Centre for Disease Control

View shared research outputs
Top Co-Authors

Avatar

Zsuzsanna Hollander

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

J. Wilson-McManus

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

David Lin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Alice Mui

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Andrew Ignaszewski

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Virginia Chen

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge