Network


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

Hotspot


Dive into the research topics where David A. Priestman is active.

Publication


Featured researches published by David A. Priestman.


Nature Genetics | 2004

Infantile-onset symptomatic epilepsy syndrome caused by a homozygous loss-of-function mutation of GM3 synthase

Michael A. Simpson; Harold E. Cross; Christos Proukakis; David A. Priestman; David C. A. Neville; Gabriele Reinkensmeier; Heng Wang; Max Wiznitzer; Kay Gurtz; Argyro Verganelaki; Anna Pryde; Michael A. Patton; Raymond A. Dwek; Terry D. Butters; Frances M. Platt; Andrew H. Crosby

We identified an autosomal recessive infantile-onset symptomatic epilepsy syndrome associated with developmental stagnation and blindness. Assuming a founder effect in a large Old Order Amish pedigree, we carried out a genome-wide screen for linkage and identified a single region of homozygosity on chromosome 2p12–p11.2 spanning 5.1 cM (maximum lod score of 6.84). We sequenced genes in the region and identified a nonsense mutation in SIAT9, which is predicted to result in the premature termination of the GM3 synthase enzyme (also called lactosylceramide α-2,3 sialyltransferase). GM3 synthase is a member of the sialyltransferase family and catalyzes the initial step in the biosynthesis of most complex gangliosides from lactosylceramide. Biochemical analysis of plasma glycosphingolipids confirmed that affected individuals lack GM3 synthase activity, as marked by a complete lack of GM3 ganglioside and its biosynthetic derivatives and an increase in lactosylceramide and its alternative derivatives. Although the relationship between defects in ganglioside catabolism and a range of lysosomal storage diseases is well documented, this is the first report, to our knowledge, of a disruption of ganglioside biosynthesis associated with human disease.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Implications for invariant natural killer T cell ligands due to the restricted presence of isoglobotrihexosylceramide in mammals

Anneliese O. Speak; Mariolina Salio; David C. A. Neville; Josette Fontaine; David A. Priestman; Nick Platt; Tanya Heare; Terry D. Butters; Raymond A. Dwek; François Trottein; Mark A. Exley; Vincenzo Cerundolo; Frances M. Platt

Development of invariant natural killer T (iNKT) cells requires the presentation of lipid ligand(s) by CD1d molecules in the thymus. The glycosphingolipid (GSL) isoglobotrihexosylceramide (iGb3) has been proposed as the natural iNKT cell-selecting ligand in the thymus and to be involved in peripheral activation of iNKT cells by dendritic cells (DCs). However, there is no direct biochemical evidence for the presence of iGb3 in mouse or human thymus or DCs. Using a highly sensitive HPLC assay, the only tissue where iGb3 could be detected in mouse was the dorsal root ganglion (DRG). iGb3 was not detected in other mouse or any human tissues analyzed, including thymus and DCs. Even in mutant mice that store isoglobo-series GSLs in the DRG, we were still unable to detect these GSLs in the thymus. iGb3 is therefore unlikely to be a physiologically relevant iNKT cell-selecting ligand in mouse and humans. A detailed study is now warranted to better understand the nature of iNKT cell-selecting ligand(s) in vivo.


Journal of the American Society for Mass Spectrometry | 2001

Ionization and Fragmentation of Neutral and Acidic Glycosphingolipids with a Q-TOF Mass Spectrometer Fitted with a MALDI Ion Source

Victoria Hunnam; David J. Harvey; David A. Priestman; Robert Harold Bateman; Robert S. Bordoli; Richard Tyldesley

This paper reports the use of a quadrupole time-of-flight (Q-TOF) mass spectrometer fitted with a matrix-assisted laser desorption/ionization (MALDI) ion source for the analysis of neutral and acidic glycosphingolipids. All compounds gave strong [M + Na]+ ions with 2,5-dihydroxybenzoic acid as the matrix, with no loss of sensitivity with increasing mass as was observed from the corresponding ions produced by electrospray. Neutral glycosphingolipids showed negligible in-source fragmentation but sialylated compounds fragmented by loss of sialic acid. However, these losses were not accompanied by unfocused post-source-decay ions as observed with MALDI-reflectron-TOF instruments. The MS/MS spectra were almost identical to those obtained by electrospray. Fragmentation of all compounds was mainly by glycosidic cleavage to give ions, both with and without the ceramide moiety, which defined the carbohydrate chain sequence. Weak ions which defined the sphingosine chain length and abundant ions, produced by loss of the acyl chain, were present when this chain contained a 2-hydroxy group. The technique was applied to the identification of ceramide— trihexosides present in tissues from mice genetically modified to model one of the glycolipid storage diseases (Fabry disease).


Brain | 2013

Mutations in B4GALNT1 (GM2 synthase) underlie a new disorder of ganglioside biosynthesis

Gaurav V. Harlalka; Anna Lehman; Barry A. Chioza; Emma L. Baple; Reza Maroofian; Harold E. Cross; Ajith Sreekantan-Nair; David A. Priestman; Saeed Al-Turki; Meriel McEntagart; Christos Proukakis; Louise Royle; Radoslaw P. Kozak; Laila Bastaki; Michael A. Patton; K. Wagner; Roselyn Coblentz; Joy Price; Michelle M. Mezei; Kamilla Schlade-Bartusiak; Frances M. Platt; Andrew H. Crosby

Glycosphingolipids are ubiquitous constituents of eukaryotic plasma membranes, and their sialylated derivatives, gangliosides, are the major class of glycoconjugates expressed by neurons. Deficiencies in their catabolic pathways give rise to a large and well-studied group of inherited disorders, the lysosomal storage diseases. Although many glycosphingolipid catabolic defects have been defined, only one proven inherited disease arising from a defect in ganglioside biosynthesis is known. This disease, because of defects in the first step of ganglioside biosynthesis (GM3 synthase), results in a severe epileptic disorder found at high frequency amongst the Old Order Amish. Here we investigated an unusual neurodegenerative phenotype, most commonly classified as a complex form of hereditary spastic paraplegia, present in families from Kuwait, Italy and the Old Order Amish. Our genetic studies identified mutations in B4GALNT1 (GM2 synthase), encoding the enzyme that catalyzes the second step in complex ganglioside biosynthesis, as the cause of this neurodegenerative phenotype. Biochemical profiling of glycosphingolipid biosynthesis confirmed a lack of GM2 in affected subjects in association with a predictable increase in levels of its precursor, GM3, a finding that will greatly facilitate diagnosis of this condition. With the description of two neurological human diseases involving defects in two sequentially acting enzymes in ganglioside biosynthesis, there is the real possibility that a previously unidentified family of ganglioside deficiency diseases exist. The study of patients and animal models of these disorders will pave the way for a greater understanding of the role gangliosides play in neuronal structure and function and provide insights into the development of effective treatment therapies.


Science Translational Medicine | 2016

Heat shock protein–based therapy as a potential candidate for treating the sphingolipidoses

Thomas Kirkegaard; James Gray; David A. Priestman; Kerri L. Wallom; Jennifer Atkins; Ole Dines Olsen; Alexander Klein; Svetlana Drndarski; Nikolaj H.T. Petersen; Linda Ingemann; David A. Smith; Lauren Morris; Claus Bornæs; Signe Humle Jørgensen; Ian M. Williams; Anders Hinsby; Christoph Arenz; David J. Begley; Marja Jäättelä; Frances M. Platt

Increasing Hsp70 expression in lysosomes using the small-molecule arimoclomol ameliorates pathology in several animal models of sphingolipidoses. Heat shock protein to the rescue The sphingolipidoses constitute a major subgroup of lysosomal storage diseases, a class of inherited metabolic disorders characterized by severe systemic and neurological problems. Few therapeutic options exist for treating these disorders. Kirkegaard et al. now demonstrate that increasing the expression of the molecular chaperone HSP70 through administration of either recombinant human HSP70 or the clinically tested, orally available small-molecule arimoclomol ameliorated disease manifestations, including brain pathology, in several different animal models of sphingolipidoses. Lysosomal storage diseases (LSDs) often manifest with severe systemic and central nervous system (CNS) symptoms. The existing treatment options are limited and have no or only modest efficacy against neurological manifestations of disease. We demonstrate that recombinant human heat shock protein 70 (HSP70) improves the binding of several sphingolipid-degrading enzymes to their essential cofactor bis(monoacyl)glycerophosphate in vitro. HSP70 treatment reversed lysosomal pathology in primary fibroblasts from 14 patients with eight different LSDs. HSP70 penetrated effectively into murine tissues including the CNS and inhibited glycosphingolipid accumulation in murine models of Fabry disease (Gla−/−), Sandhoff disease (Hexb−/−), and Niemann-Pick disease type C (Npc1−/−) and attenuated a wide spectrum of disease-associated neurological symptoms in Hexb−/− and Npc1−/− mice. Oral administration of arimoclomol, a small-molecule coinducer of HSPs that is currently in clinical trials for Niemann-Pick disease type C (NPC), recapitulated the effects of recombinant human HSP70, suggesting that heat shock protein–based therapies merit clinical evaluation for treating LSDs.


FEBS Letters | 1992

Purification and partial characterization of rat liver pyruvate dehydrogenase kinase activator protein (free pyruvate dehydrogenase kinase)

David A. Priestman; Sharad C. Mistry; Alan L. Kerbey; Philip J. Randle

Rat liver pyruvate dehydrogenase (PDH) kinase activator protein (KAP), a free PDH kinase readily separable from PDH complex and its intrinsic kinase, has been purified to apparent homogeneity from liver mitochondria of fed and 48‐h starved rats. On SDS‐PAGE an apparently single band of M, 45 kDa was obtained. N‐Terminal amino acid sequence analyses (8–10 cycles) confirmed the presence of a single peptide in each case. The specific activity of the purified KAP from 48‐h starved rats (14,413 U/mg protein) was 4.5‐fold greater than that from fed rats.


Human Molecular Genetics | 2015

Amyotrophic lateral sclerosis and denervation alter sphingolipids and up-regulate glucosylceramide synthase

Alexandre Henriques; Vincent Croixmarie; David A. Priestman; Angela Rosenbohm; Sylvie Dirrig-Grosch; Eleonora D'Ambra; Mylene Huebecker; Ghulam Hussain; Claire Boursier-Neyret; Andoni Echaniz-Laguna; Albert C. Ludolph; Frances M. Platt; Bernard Walther; Michael Spedding; Jean-Philippe Loeffler; Jose-Luis Gonzalez de Aguilar

Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset disease characterized by upper and lower motor neuron degeneration, muscle wasting and paralysis. Growing evidence suggests a link between changes in lipid metabolism and ALS. Here, we used UPLC/TOF-MS to survey the lipidome in SOD1(G86R) mice, a model of ALS. Significant changes in lipid expression were evident in spinal cord and skeletal muscle before overt neuropathology. In silico analysis also revealed appreciable changes in sphingolipids including ceramides and glucosylceramides (GlcCer). HPLC analysis showed increased amounts of GlcCer and downstream glycosphingolipids (GSLs) in SOD1(G86R) muscle compared with wild-type littermates. Glucosylceramide synthase (GCS), the enzyme responsible for GlcCer biosynthesis, was up-regulated in muscle of SOD1(G86R) mice and ALS patients, and in muscle of wild-type mice after surgically induced denervation. Conversely, inhibition of GCS in wild-type mice, following transient peripheral nerve injury, reversed the overexpression of genes in muscle involved in oxidative metabolism and delayed motor recovery. GCS inhibition in SOD1(G86R) mice also affected the expression of metabolic genes and induced a loss of muscle strength and morphological deterioration of the motor endplates. These findings suggest that GSLs may play a critical role in ALS muscle pathology and could lead to the identification of new therapeutic targets.


Journal of Biological Chemistry | 2014

A Novel Mouse Model of a Patient Mucolipidosis II Mutation Recapitulates Disease Pathology

Leigh Paton; Emmanuelle Bitoun; Janet Kenyon; David A. Priestman; Peter L. Oliver; Benjamin Edwards; Frances M. Platt; Kay E. Davies

Background: Mucolipidosis II is a severe lysosomal storage disorder, fatal in childhood and lacking drug treatments. Results: This novel mouse model of a mucolipidosis II patient mutation recapitulates the human pathology. Conclusion: Mouse models based on patient mutations are more valuable to study mucolipidosis II than a knock-out of the gene. Significance: This novel mouse model will be useful for future drug development. Mucolipidosis II (MLII) is a lysosomal storage disorder caused by loss of N-acetylglucosamine-1-phosphotransferase, which tags lysosomal enzymes with a mannose 6-phosphate marker for transport to the lysosome. In MLII, the loss of this marker leads to deficiency of multiple enzymes and non-enzymatic proteins in the lysosome, leading to the storage of multiple substrates. Here we present a novel mouse model of MLII homozygous for a patient mutation in the GNPTAB gene. Whereas the current gene knock-out mouse model of MLII lacks some of the characteristic features of the human disease, our novel mouse model more fully recapitulates the human pathology, showing growth retardation, skeletal and facial abnormalities, increased circulating lysosomal enzymatic activities, intracellular lysosomal storage, and reduced life span. Importantly, MLII behavioral deficits are characterized for the first time, including impaired motor function and psychomotor retardation. Histological analysis of the brain revealed progressive neurodegeneration in the cerebellum with severe Purkinje cell loss as the underlying cause of the ataxic gait. In addition, based on the loss of Npc2 (Niemann-Pick type C 2) protein expression in the brain, the mice were treated with 2-hydroxypropyl-β-cyclodextrin, a drug previously reported to rescue Purkinje cell death in a mouse model of Niemann-Pick type C disease. No improvement in brain pathology was observed. This indicates that cerebellar degeneration is not primarily triggered by loss of Npc2 function. This study emphasizes the value of modeling MLII patient mutations to generate clinically relevant mouse mutants to elucidate the pathogenic molecular pathways of MLII and address their amenability to therapy.


Scientific Reports | 2017

Inhibition of β-Glucocerebrosidase Activity Preserves Motor Unit Integrity in a Mouse Model of Amyotrophic Lateral Sclerosis.

Alexandre Henriques; Mylene Huebecker; Hélène Blasco; Céline Keime; Christian R. Andres; Philippe Corcia; David A. Priestman; Frances M. Platt; Michael Spedding; Jean-Philippe Loeffler

Recent metabolomic reports connect dysregulation of glycosphingolipids, particularly ceramide and glucosylceramide, to neurodegeneration and to motor unit dismantling in amyotrophic lateral sclerosis at late disease stage. We report here altered levels of gangliosides in the cerebrospinal fluid of amyotrophic lateral sclerosis patients in early disease stage. Conduritol B epoxide is an inhibitor of acid beta-glucosidase, and lowers glucosylceramide degradation. Glucosylceramide is the precursor for all of the more complex glycosphingolipids. In SOD1G86R mice, an animal model of amyotrophic lateral sclerosis, conduritol B epoxide preserved ganglioside distribution at the neuromuscular junction, delayed disease onset, improved motor function and preserved motor neurons as well as neuromuscular junctions from degeneration. Conduritol B epoxide mitigated gene dysregulation in the spinal cord and restored the expression of genes involved in signal transduction and axonal elongation. Inhibition of acid beta-glucosidase promoted faster axonal elongation in an in vitro model of neuromuscular junctions and hastened recovery after peripheral nerve injury in wild type mice. Here, we provide evidence that glycosphingolipids play an important role in muscle innervation, which degenerates in amyotrophic lateral sclerosis from the early disease stage. This is a first proof of concept study showing that modulating the catabolism of glucosylceramide may be a therapeutic target for this devastating disease.


Nature Medicine | 2018

Fetal gene therapy for neurodegenerative disease of infants.

Giulia Massaro; Citra Nurfarah Zaini Mattar; Andrew Wong; Ernestas Sirka; Suzanne M. K. Buckley; Bronwen R. Herbert; Stefan Karlsson; Dany Perocheau; Derek Burke; Simon Heales; Angela Richard-Londt; Sebastian Brandner; Mylene Huebecker; David A. Priestman; Frances M. Platt; Kevin Mills; Arijit Biswas; Jonathan D. Cooper; Jerry Chan; Seng H. Cheng; Simon N. Waddington; Ahad A. Rahim

For inherited genetic diseases, fetal gene therapy offers the potential of prophylaxis against early, irreversible and lethal pathological change. To explore this, we studied neuronopathic Gaucher disease (nGD), caused by mutations in GBA. In adult patients, the milder form presents with hepatomegaly, splenomegaly and occasional lung and bone disease; this is managed, symptomatically, by enzyme replacement therapy. The acute childhood lethal form of nGD is untreatable since enzyme cannot cross the blood–brain barrier. Patients with nGD exhibit signs consistent with hindbrain neurodegeneration, including neck hyperextension, strabismus and, often, fatal apnea1. We selected a mouse model of nGD carrying a loxP-flanked neomycin disruption of Gba plus Cre recombinase regulated by the keratinocyte-specific K14 promoter. Exclusive skin expression of Gba prevents fatal neonatal dehydration. Instead, mice develop fatal neurodegeneration within 15 days2. Using this model, fetal intracranial injection of adeno-associated virus (AAV) vector reconstituted neuronal glucocerebrosidase expression. Mice lived for up to at least 18 weeks, were fertile and fully mobile. Neurodegeneration was abolished and neuroinflammation ameliorated. Neonatal intervention also rescued mice but less effectively. As the next step to clinical translation, we also demonstrated the feasibility of ultrasound-guided global AAV gene transfer to fetal macaque brains.In utero GBA gene therapy extends lifespan and provides long-lasting phenotypic amelioration in a mouse model of neuronopathic Gaucher disease. Fetal ultrasound-guided in utero gene vector delivery is also achieved in the non-human primate brain.

Collaboration


Dive into the David A. Priestman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David J. Harvey

University College Dublin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge