Network


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

Hotspot


Dive into the research topics where Douglas Macdonald is active.

Publication


Featured researches published by Douglas Macdonald.


Journal of Clinical Investigation | 2012

Mutant huntingtin fragmentation in immune cells tracks Huntington’s disease progression

Andreas Weiss; Ulrike Träger; Edward J. Wild; Stephan Grueninger; Ruth Farmer; Christian Landles; Rachael I. Scahill; Nayana Lahiri; Salman Haider; Douglas Macdonald; Chris Frost; Gillian P. Bates; Graeme Bilbe; Rainer Kuhn; Ralph Andre; Sarah J. Tabrizi

Huntingtons disease (HD) is a fatal, inherited neurodegenerative disorder caused by an expanded CAG repeat in the gene encoding huntingtin (HTT). Therapeutic approaches to lower mutant HTT (mHTT) levels are expected to proceed to human trials, but noninvasive quantification of mHTT is not currently possible. The importance of the peripheral immune system in neurodegenerative disease is becoming increasingly recognized. Peripheral immune cells have been implicated in HD pathogenesis, but HTT levels in these cells have not been quantified before. A recently described time-resolved Förster resonance energy transfer (TR-FRET) immunoassay was used to quantify mutant and total HTT protein levels in leukocytes from patients with HD. Mean mHTT levels in monocytes, T cells, and B cells differed significantly between patients with HD and controls and between pre-manifest mutation carriers and those with clinical onset. Monocyte and T cell mHTT levels were significantly associated with disease burden scores and caudate atrophy rates in patients with HD. mHTT N-terminal fragments detected in HD PBMCs may explain the progressive increase in mHTT levels in these cells. These findings indicate that quantification of mHTT in peripheral immune cells by TR-FRET holds significant promise as a noninvasive disease biomarker.


Journal of Clinical Investigation | 2015

Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington’s disease patients

Edward J. Wild; Roberto Boggio; Douglas R. Langbehn; Nicola J. Robertson; Salman Haider; James R. Miller; Henrik Zetterberg; Blair R. Leavitt; Rainer Kuhn; Sarah J. Tabrizi; Douglas Macdonald; Andreas Weiss

BACKGROUNDnQuantification of disease-associated proteins in the cerebrospinal fluid (CSF) has been critical for the study and treatment of several neurodegenerative disorders; however, mutant huntingtin protein (mHTT), the cause of Huntingtons disease (HD), is at very low levels in CSF and, to our knowledge, has never been measured previously.nnnMETHODSnWe developed an ultrasensitive single-molecule counting (SMC) mHTT immunoassay that was used to quantify mHTT levels in CSF samples from individuals bearing the HD mutation and from control individuals in 2 independent cohorts.nnnRESULTSnThis SMC mHTT immunoassay demonstrated high specificity for mHTT, high sensitivity with a femtomolar detection threshold, and a broad dynamic range. Analysis of the CSF samples showed that mHTT was undetectable in CSF from all controls but quantifiable in nearly all mutation carriers. The mHTT concentration in CSF was approximately 3-fold higher in patients with manifest HD than in premanifest mutation carriers. Moreover, mHTT levels increased as the disease progressed and were associated with 5-year onset probability. The mHTT concentration independently predicted cognitive and motor dysfunction. Furthermore, the level of mHTT was associated with the concentrations of tau and neurofilament light chain in the CSF, suggesting a neuronal origin for the detected mHTT.nnnCONCLUSIONSnWe have demonstrated that mHTT can be quantified in CSF from HD patients using the described SMC mHTT immunoassay. Moreover, the level of mHTT detected is associated with proximity to disease onset and diminished cognitive and motor function. The ability to quantify CSF mHTT will facilitate the study of HD, and mHTT quantification could potentially serve as a biomarker for the development and testing of experimental mHTT-lowering therapies for HD.nnnTRIAL REGISTRATIONnNot applicable.nnnFUNDINGnCHDI Foundation Inc.; Medical Research Council (MRC) UK; National Institutes for Health Research (NIHR); Rosetrees Trust; Swedish Research Council; and Knut and Alice Wallenberg Foundation.


Journal of Biomolecular Screening | 2010

A Profiling Platform for the Characterization of Transglutaminase 2 (TG2) Inhibitors

Sabine Schaertl; Michael Prime; John Wityak; Celia Dominguez; Ignacio Munoz-Sanjuan; Robert Pacifici; Stephen Martin Courtney; Andreas Scheel; Douglas Macdonald

Huntington’s disease (HD) is associated with increased expression levels and activity of tissue transglutaminase (TG2), an enzyme primarily known for its cross-linking of proteins. To validate TG2 as a therapeutic target for HD in transgenic models and for eventual clinical development, a selective and brain-permeable inhibitor is required. Here, a comprehensive profiling platform of biochemical and cellular assays is presented which has been established to evaluate the potency, cellular efficacy, subtype selectivity and the mechanism-of-action of known and novel TG2 inhibitors. Several classes of inhibitors have been characterized including: the commonly used pseudo-substrate inhibitors, cystamine and putrescine (which are generally nonspecific for TG2 and therefore not practical for drug development), the various peptidic inhibitors that target the active site cysteine residue (which display excellent selectivity but in general have poor cellular activity), and the allosteric reversible small-molecule hydrazides (which show poor selectivity and a lack of cellular activity and could not be improved despite considerable medicinal chemistry efforts). In addition, a set of inhibitors identified from a collection of pharmacologically active compounds was found to be unselective for TG2. Moreover, inhibition at the guanosine triphosphate binding site has been examined, but apart from guanine nucleotides, no such inhibitors have been identified. In addition, the promising pharmacological profile of a TG2 inhibitor is presented which is currently in lead optimization to be developed as a tool compound.


Scientific Reports | 2016

A novel, native-format bispecific antibody triggering T-cell killing of B-cells is robustly active in mouse tumor models and cynomolgus monkeys

Eric Smith; Kara Olson; Lauric Haber; Bindu Varghese; Paurene Duramad; Andrew D. Tustian; Adelekan Oyejide; Jessica R. Kirshner; Lauren Canova; Jayanthi Menon; Jennifer Principio; Douglas Macdonald; Joel Kantrowitz; Nicholas J. Papadopoulos; Neil Stahl; George D. Yancopoulos; Gavin Thurston; Samuel Davis

Bispecific antibodies, while showing great therapeutic potential, pose formidable challenges with respect to their assembly, stability, immunogenicity, and pharmacodynamics. Here we describe a novel class of bispecific antibodies with native human immunoglobulin format. The design exploits differences in the affinities of the immunoglobulin isotypes for Protein A, allowing efficient large-scale purification. Using this format, we generated a bispecific antibody, REGN1979, targeting the B cell marker, CD20, and the CD3 component of the T cell receptor, which triggers redirected killing of B cells. In mice, this antibody prevented growth of B cell tumors and also caused regression of large established tumors. In cynomolgus monkeys, low doses of REGN1979 caused prolonged depletion of B cells in peripheral blood with a serum half-life of approximately 14 days. Further, the antibody induced a deeper depletion of B cells in lymphoid organs than rituximab. This format has broad applicability for development of clinical bispecific antibodies.


Journal of Medicinal Chemistry | 2012

Discovery and Structure–Activity Relationship of Potent and Selective Covalent Inhibitors of Transglutaminase 2 for Huntington’s Disease

Michael Prime; Ole Andreas Andersen; John J. Barker; Mark Brooks; Robert K. Y. Cheng; Ian Toogood-Johnson; Stephen Martin Courtney; Frederick Arthur Brookfield; Christopher John Yarnold; Richard W. Marston; Peter Johnson; Siw Johnsen; Jordan J. Palfrey; Darshan Vaidya; Sayeh Erfan; Osamu Ichihara; Brunella Felicetti; Shilpa Palan; Anna Pedret-Dunn; Sabine Schaertl; Ina Sternberger; Andreas Ebneth; Andreas Scheel; Dirk Winkler; Leticia Toledo-Sherman; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez; John Wityak

Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntingtons disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.


Amino Acids | 2012

TG2 transamidating activity acts as a reostat controlling the interplay between apoptosis and autophagy

Federica Rossin; Manuela D'Eletto; Douglas Macdonald; Maria Grazia Farrace; Mauro Piacentini

Tissue transglutaminase (TG2) activity has been implicated in inflammatory disease processes such as Celiac disease, infectious diseases, cancer, and neurodegenerative diseases, such as Huntington’s disease. Furthermore, four distinct biochemical activities have been described for TG2 including protein crosslinking via transamidation, GTPase, kinase and protein disulfide isomerase activities. Although the enzyme plays a complex role in the regulation of cell death and autophagy, the molecular mechanisms and the putative biochemical activity involved in each is unclear. Therefore, the goal of the present study was to determine how TG2 modulates autophagy and/or apoptosis and which of its biochemical activities is involved in those processes. To address this question, immortalized embryonic fibroblasts obtained from TG2 knock-out mice were reconstituted with either wild-type TG2 or TG2 lacking its transamidating activity and these were subjected to different treatments to induce autophagy or apoptosis. We found that knock out of the endogenous TG2 resulted in a significant exacerbation of caspase 3 activity and PARP cleavage in MEF cells subjected to apoptotic stimuli. Interestingly, the same cells showed the accumulation of LC3 II isoform following autophagy induction. These findings strongly suggest that TG2 transamidating activity plays a protective role in the response of MEF cells to death stimuli, because the expression of the wild-type TG2, but not its transamidation inactive C277S mutant, resulted in a suppression of caspase 3 as well as PARP cleavage upon apoptosis induction. Additionally, the same mutant was unable to catalyze the final steps in autophagosome formation during autophagy. Our findings clearly indicate that the TG2 transamidating activity is the primary biochemical function involved in the physiological regulation of both apoptosis and autophagy. These data also indicate that TG2 is a key regulator of cross-talk between autophagy and apoptosis.


PLOS ONE | 2014

Polyglutamine- and Temperature-Dependent Conformational Rigidity in Mutant Huntingtin Revealed by Immunoassays and Circular Dichroism Spectroscopy

Valentina Fodale; Natalie C. Kegulian; Margherita Verani; Cristina Cariulo; Lucia Azzollini; Lara Petricca; Manuel Daldin; Roberto Boggio; Alessandro Padova; Rainer Kuhn; Robert Pacifici; Douglas Macdonald; Ryan C. Schoenfeld; Hyunsun Park; J. Mario Isas; Ralf Langen; Andreas Weiss; Andrea Caricasole

Background In Huntingtons disease, expansion of a CAG triplet repeat occurs in exon 1 of the huntingtin gene (HTT), resulting in a protein bearing>35 polyglutamine residues whose N-terminal fragments display a high propensity to misfold and aggregate. Recent data demonstrate that polyglutamine expansion results in conformational changes in the huntingtin protein (HTT), which likely influence its biological and biophysical properties. Developing assays to characterize and measure these conformational changes in isolated proteins and biological samples would advance the testing of novel therapeutic approaches aimed at correcting mutant HTT misfolding. Time-resolved Förster energy transfer (TR-FRET)-based assays represent high-throughput, homogeneous, sensitive immunoassays widely employed for the quantification of proteins of interest. TR-FRET is extremely sensitive to small distances and can therefore provide conformational information based on detection of exposure and relative position of epitopes present on the target protein as recognized by selective antibodies. We have previously reported TR-FRET assays to quantify HTT proteins based on the use of antibodies specific for different amino-terminal HTT epitopes. Here, we investigate the possibility of interrogating HTT protein conformation using these assays. Methodology/Principal Findings By performing TR-FRET measurements on the same samples (purified recombinant proteins or lysates from cells expressing HTT fragments or full length protein) at different temperatures, we have discovered a temperature-dependent, reversible, polyglutamine-dependent conformational change of wild type and expanded mutant HTT proteins. Circular dichroism spectroscopy confirms the temperature and polyglutamine-dependent change in HTT structure, revealing an effect of polyglutamine length and of temperature on the alpha-helical content of the protein. Conclusions/Significance The temperature- and polyglutamine-dependent effects observed with TR-FRET on HTT proteins represent a simple, scalable, quantitative and sensitive assay to identify genetic and pharmacological modulators of mutant HTT conformation, and potentially to assess the relevance of conformational changes during onset and progression of Huntingtons disease.


PLOS ONE | 2014

Quantification Assays for Total and Polyglutamine-Expanded Huntingtin Proteins

Douglas Macdonald; Michela Tessari; Ivette Boogaard; Melanie Smith; Kristiina Pulli; Agnieszka Szynol; Faywell Albertus; Marieke B.A.C. Lamers; Sipke Dijkstra; Daniel Kordt; Wolfgang Reindl; Frank Herrmann; George McAllister; David F. Fischer; Ignacio Munoz-Sanjuan

The expansion of a CAG trinucleotide repeat in the huntingtin gene, which produces huntingtin protein with an expanded polyglutamine tract, is the cause of Huntingtons disease (HD). Recent studies have reported that RNAi suppression of polyglutamine-expanded huntingtin (mutant HTT) in HD animal models can ameliorate disease phenotypes. A key requirement for such preclinical studies, as well as eventual clinical trials, aimed to reduce mutant HTT exposure is a robust method to measure HTT protein levels in select tissues. We have developed several sensitive and selective assays that measure either total human HTT or polyglutamine-expanded human HTT proteins on the electrochemiluminescence Meso Scale Discovery detection platform with an increased dynamic range over other methods. In addition, we have developed an assay to detect endogenous mouse and rat HTT proteins in pre-clinical models of HD to monitor effects on the wild type protein of both allele selective and non-selective interventions. We demonstrate the application of these assays to measure HTT protein in several HD in vitro cellular and in vivo animal model systems as well as in HD patient biosamples. Furthermore, we used purified recombinant HTT proteins as standards to quantitate the absolute amount of HTT protein in such biosamples.


ACS Medicinal Chemistry Letters | 2012

SAR Development of Lysine-Based Irreversible Inhibitors of Transglutaminase 2 for Huntington's Disease

John Wityak; Michael Prime; Frederick Arthur Brookfield; Stephen Martin Courtney; Sayeh Erfan; Siw Johnsen; Peter Johnson; Marie Li; Richard W. Marston; Laura Reed; Darshan Vaidya; Sabine Schaertl; Anna Pedret-Dunn; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez

We report a series of irreversible transglutaminase 2 inhibitors starting from a known lysine dipeptide bearing an acrylamide warhead. We established new SARs resulting in compounds demonstrating improved potency and better physical and calculated properties. Transglutaminase selectivity profiling and in vitro ADME properties of selected compounds are also reported.


ACS Medicinal Chemistry Letters | 2012

Irreversible 4-Aminopiperidine Transglutaminase 2 Inhibitors for Huntington's Disease.

Michael Prime; Frederick Arthur Brookfield; Stephen Martin Courtney; Simon Gaines; Richard W. Marston; Osamu Ichihara; Marie Li; Darshan Vaidya; Helen Williams; Anna Pedret-Dunn; Laura Reed; Sabine Schaertl; Leticia Toledo-Sherman; Maria Beconi; Douglas Macdonald; Ignacio Munoz-Sanjuan; Celia Dominguez; John Wityak

A new series of potent TG2 inhibitors are reported that employ a 4-aminopiperidine core bearing an acrylamide warhead. We establish the structure-activity relationship of this new series and report on the transglutaminase selectivity and in vitro ADME properties of selected compounds. We demonstrate that the compounds do not conjugate glutathione in an in vitro setting and have superior plasma stability over our previous series.

Collaboration


Dive into the Douglas Macdonald's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Wityak

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah J. Tabrizi

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge