Network


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

Hotspot


Dive into the research topics where Angelique R. Ormsby is active.

Publication


Featured researches published by Angelique R. Ormsby.


Nature Methods | 2012

Tracking protein aggregation and mislocalization in cells with flow cytometry

Yasmin M. Ramdzan; Saskia Polling; Cheryl Chia; Ivan Ng; Angelique R. Ormsby; Nathan P. Croft; Anthony W. Purcell; Marie A. Bogoyevitch; Dominic C. H. Ng; Paul A. Gleeson; Danny M. Hatters

We applied pulse-shape analysis (PulSA) to monitor protein localization changes in mammalian cells by flow cytometry. PulSA enabled high-throughput tracking of protein aggregation, translocation from the cytoplasm to the nucleus and trafficking from the plasma membrane to the Golgi as well as stress-granule formation. Combining PulSA with tetracysteine-based oligomer sensors in a cell model of Huntingtons disease enabled further separation of cells enriched with monomers, oligomers and inclusion bodies.


Cell Reports | 2017

Huntingtin Inclusions Trigger Cellular Quiescence, Deactivate Apoptosis, and Lead to Delayed Necrosis

Yasmin M. Ramdzan; Mikhail M. Trubetskov; Angelique R. Ormsby; Estella A. Newcombe; Xiaojing Sui; Mark J. Tobin; Marie N. Bongiovanni; Sally L. Gras; Grant Dewson; Jason Miller; Steven Finkbeiner; Nagaraj S. Moily; Jonathan C. Niclis; Clare L. Parish; Anthony W. Purcell; Michael J. Baker; Jacqueline A. Wilce; Saboora Waris; Diana Stojanovski; Till Böcking; Ching-Seng Ang; David B. Ascher; Gavin E. Reid; Danny M. Hatters

Competing models exist in the literature for the relationship between mutant Huntingtin exon 1 (Httex1) inclusion formation and toxicity. In one, inclusions are adaptive by sequestering the proteotoxicity of soluble Httex1. In the other, inclusions compromise cellular activity as a result of proteome co-aggregation. Using a biosensor of Httex1 conformation in mammalian cell models, we discovered a mechanism that reconciles these competing models. Newly formed inclusions were composed of disordered Httex1 and ribonucleoproteins. As inclusions matured, Httex1 reconfigured into amyloid, and other glutamine-rich and prion domain-containing proteins were recruited. Soluble Httex1 caused a hyperpolarized mitochondrial membrane potential, increased reactive oxygen species, and promoted apoptosis. Inclusion formation triggered a collapsed mitochondrial potential, cellular quiescence, and deactivated apoptosis. We propose a revised model where sequestration of soluble Httex1 inclusions can remove the trigger for apoptosis but also co-aggregate other proteins, which curtails cellular metabolism and leads to a slow death by necrosis.


Journal of Biological Chemistry | 2013

A Platform to View Huntingtin Exon 1 Aggregation Flux in the Cell Reveals Divergent Influences from Chaperones hsp40 and hsp70

Angelique R. Ormsby; Yasmin M. Ramdzan; Yee-Foong Mok; Kristijan D. Jovanoski; Danny M. Hatters

Background: How misfolded proteins such as mutant huntingtin aggregate in the cell remains enigmatic. Results: We built a platform to view how aggregation proceeds and assessed the impact of quality control chaperones hsp40 and hsp70. Conclusion: hsp70 enhanced survival of cells with aggregates; hsp40 suppressed aggregation. Significance: We developed a new toolkit to illustrate the impact of protein aggregation on cell biology. Our capacity for tracking how misfolded proteins aggregate inside a cell and how different aggregation states impact cell biology remains enigmatic. To address this, we built a new toolkit that enabled the high throughput tracking of individual cells enriched with polyglutamine-expanded Htt exon 1 (Httex1) monomers, oligomers, and inclusions using biosensors of aggregation state and flow cytometry pulse shape analysis. Supplemented with gel filtration chromatography and fluorescence-adapted sedimentation velocity analysis of cell lysates, we collated a multidimensional view of Httex1 aggregation in cells with respect to time, polyglutamine length, expression levels, cell survival, and overexpression of protein quality control chaperones hsp40 (DNAJB1) and hsp70 (HSPA1A). Cell death rates trended higher for Neuro2a cells containing Httex1 in inclusions than with Httex1 dispersed through the cytosol at time points of expression over 2 days. hsp40 stabilized monomers and suppressed inclusion formation but did not otherwise change Httex1 toxicity. hsp70, however, had no major effect on aggregation of Httex1 but increased the survival rate of cells with inclusions. hsp40 and hsp70 also increased levels of a second bicistronic reporter of Httex1 expression, mKate2, and increased total numbers of cells in culture, suggesting these chaperones partly rectify Httex1-induced deficiencies in quality control and growth rates. Collectively, these data suggest that Httex1 overstretches the protein quality control resources and that the defects can be partly rescued by overexpression of hsp40 and hsp70. Importantly, these effects occurred in a pronounced manner for soluble Httex1, which points to Httex1 aggregation occurring subsequently to more acute impacts on the cell.


PLOS ONE | 2013

Tyrosine 416 Is Phosphorylated in the Closed, Repressed Conformation of c-Src

Sevgi Irtegun; Rebecca Wood; Angelique R. Ormsby; Terrence D. Mulhern; Danny M. Hatters

c-Src kinase activity is regulated by phosphorylation of Y527 and Y416. Y527 phosphorylation stabilizes a closed conformation, which suppresses kinase activity towards substrates, whereas phosphorylation at Y416 promotes an elevated kinase activity by stabilizing the activation loop in a manner permissive for substrate binding. Here we investigated the correlation of Y416 phosphorylation with c-Src activity when c-Src was locked into the open and closed conformations (by mutations Y527F and Q528E, P529E, G530I respectively). Consistent with prior findings, we found Y416 to be more greatly phosphorylated when c-Src was in an open, active conformation. However, we also observed an appreciable amount of Y416 was phosphorylated when c-Src was in a closed, repressed conformation under conditions by which c-Src was unable to phosphorylate substrate STAT3. The phosphorylation of Y416 in the closed conformation arose by autophosphorylation, since abolishing kinase activity by mutating the ATP binding site (K295M) prevented phosphorylation. Basal Y416 phosphorylation correlated positively with cellular levels of c-Src suggesting autophosphorylation depended on self-association. Using sedimentation velocity analysis on cell lysate with fluorescence detection optics, we confirmed that c-Src forms monomers and dimers, with the open conformation also forming a minor population of larger mass complexes. Collectively, our studies suggest a model by which dimerization of c-Src primes c-Src via Y416 phosphorylation to enable rapid potentiation of activity when Src adopts an open conformation. Once in the open conformation, c-Src can amplify the response by recruiting and phosphorylating substrates such as STAT3 and increasing the extent of autophosphorylation.


Nature Communications | 2018

A biosensor-based framework to measure latent proteostasis capacity

Rebecca Wood; Angelique R. Ormsby; Mona Radwan; Dezerae Cox; Abhishek Sharma; Tobias Vöpel; Simon Ebbinghaus; Mikael Oliveberg; Gavin E. Reid; Alex Dickson; Danny M. Hatters

The pool of quality control proteins (QC) that maintains protein-folding homeostasis (proteostasis) is dynamic but can become depleted in human disease. A challenge has been in quantitatively defining the depth of the QC pool. With a new biosensor, flow cytometry-based methods and mathematical modeling we measure the QC capacity to act as holdases and suppress biosensor aggregation. The biosensor system comprises a series of barnase kernels with differing folding stability that engage primarily with HSP70 and HSP90 family proteins. Conditions of proteostasis stimulation and stress alter QC holdase activity and aggregation rates. The method reveals the HSP70 chaperone cycle to be rate limited by HSP70 holdase activity under normal conditions, but this is overcome by increasing levels of the BAG1 nucleotide exchange factor to HSPA1A or activation of the heat shock gene cluster by HSF1 overexpression. This scheme opens new paths for biosensors of disease and proteostasis systems.A pool of quality control proteins (QC) maintains the protein-folding homeostasis in the cell, but its quantitative analysis is challenging. Here the authors develop a FRET sensor based on the protein barnase, able to quantify QC holdase activity and its ability to suppress protein aggregation.


Molecular and Cellular Neuroscience | 2017

Transcriptional profiles for distinct aggregation states of mutant Huntingtin exon 1 protein unmask new Huntington's disease pathways

Nagaraj S. Moily; Angelique R. Ormsby; Aleksandar Stojilovic; Yasmin M. Ramdzan; Jeannine Diesch; Ross D. Hannan; Michelle S. Zajac; Anthony J. Hannan; Alicia Oshlack; Danny M. Hatters

&NA; Huntingtons disease is caused by polyglutamine (polyQ)‐expansion mutations in the CAG tandem repeat of the Huntingtin gene. The central feature of Huntingtons disease pathology is the aggregation of mutant Huntingtin (Htt) protein into micrometer‐sized inclusion bodies. Soluble mutant Htt states are most proteotoxic and trigger an enhanced risk of death whereas inclusions confer different changes to cellular health, and may even provide adaptive responses to stress. Yet the molecular mechanisms underpinning these changes remain unclear. Using the flow cytometry method of pulse‐shape analysis (PulSA) to sort neuroblastoma (Neuro2a) cells enriched with mutant or wild‐type Htt into different aggregation states, we clarified which transcriptional signatures were specifically attributable to cells before versus after inclusion assembly. Dampened CREB signalling was the most striking change overall and invoked specifically by soluble mutant Httex1 states. Toxicity could be rescued by stimulation of CREB signalling. Other biological processes mapped to different changes before and after aggregation included NF‐kB signalling, autophagy, SUMOylation, transcription regulation by histone deacetylases and BRD4, NAD + biosynthesis, ribosome biogenesis and altered HIF‐1 signalling. These findings open the path for therapeutic strategies targeting key molecular changes invoked prior to, and subsequently to, Httex1 aggregation. HighlightsTranscriptional changes due to aggregation of mutant Httex1 are described.The largest changes in the transcriptome are stimulated by soluble mutant Httex1.Inactivated CREB signalling is the most profound impact arising from soluble Httex1.Dampened CREB signalling explains the molecular basis of toxicity.Additional pathways unearthed as transcriptional signatures for Httex1 aggregation.


bioRxiv | 2017

Tadpole-like conformations of huntingtin exon 1 with expanded polyglutamine engenders novel interactions in cells

Estella A. Newcombe; Kiersten M. Ruff; Ashish Sethi; Angelique R. Ormsby; Yasmin M. Ramdzan; Archa H. Fox; Anthony W. Purcell; Paul R. Gooley; Rohit V. Pappu; Danny M. Hatters

Soluble huntingtin exon 1 (Httex1) with expanded polyglutamine (polyQ) engenders neurotoxicity in Huntington’s disease. To uncover the physical basis of this toxicity, we performed structural studies of soluble Httex1 for wild type and mutant polyQ lengths. Nuclear magnetic resonance experiments show evidence for conformational rigidity across the polyQ region. In contrast, hydrogen-deuterium exchange shows absence of backbone amide protection, suggesting negligible persistence of hydrogen bonds. The seemingly conflicting results are explained by all-atom simulations, which show that Httex1 adopts tadpole-like structures with a globular head encompassing the N-terminal amphipathic and polyQ regions and the tail encompassing the C-terminal proline-rich region. The surface area of the globular domain increases monotonically with polyQ length. This stimulates sharp increases in gain-of-function interactions in cells for expanded polyQ, and one of these interactions is with the stress-granule protein Fus. Our results highlight plausible connections between Httex1 structure and routes to neurotoxicity.


Journal of Huntington's disease | 2017

N-Terminal Fragments of Huntingtin Longer than Residue 170 form Visible Aggregates Independently to Polyglutamine Expansion.

Moore Z. Chen; Sue-Ann Mok; Angelique R. Ormsby; Paul J. Muchowski; Danny M. Hatters

BACKGROUND A hallmark of Huntingtons disease is the progressive aggregation of full length and N-terminal fragments of polyglutamine (polyQ)-expanded Huntingtin (Htt) into intracellular inclusions. The production of N-terminal fragments appears important for enabling pathology and aggregation; and hence the direct expression of a variety of N-terminal fragments are commonly used to model HD in animal and cellular models. OBJECTIVE It remains unclear how the length of the N-terminal fragments relates to polyQ - mediated aggregation. We investigated the fundamental intracellular aggregation process of eight different-length N-terminal fragments of Htt in both short (25Q) and long polyQ (97Q). METHODS N-terminal fragments were fused to fluorescent proteins and transiently expressed in mammalian cell culture models. These included the classic exon 1 fragment (90 amino acids) and longer forms of 105, 117, 171, 513, 536, 552, and 586 amino acids based on wild-type Htt (of 23Q) sequence length nomenclature. RESULTS N-terminal fragments of less than 171 amino acids only formed inclusions in polyQ-expanded form. By contrast the longer fragments formed inclusions irrespective of Q-length, with Q-length playing a negligible role in extent of aggregation. The inclusions could be classified into 3 distinct morphological categories. One type (Type A) was universally associated with polyQ expansions whereas the other two types (Types B and C) formed independently of polyQ length expansion. CONCLUSIONS PolyQ-expansion was only required for fragments of less than 171 amino acids to aggregate. Longer fragments aggregated predominately through a non-polyQ mechanism, involving at least one, and probably more distinct clustering mechanisms.


Journal of Molecular Biology | 2018

Tadpole-like Conformations of Huntingtin Exon 1 Are Characterized by Conformational Heterogeneity that Persists regardless of Polyglutamine Length

Estella A. Newcombe; Kiersten M. Ruff; Ashish Sethi; Angelique R. Ormsby; Yasmin M. Ramdzan; Archa H. Fox; Anthony W. Purcell; Paul R. Gooley; Rohit V. Pappu; Danny M. Hatters

Soluble huntingtin exon 1 (Httex1) with expanded polyglutamine (polyQ) engenders neurotoxicity in Huntingtons disease. To uncover the physical basis of this toxicity, we performed structural studies of soluble Httex1 for wild-type and mutant polyQ lengths. Nuclear magnetic resonance experiments show evidence for conformational rigidity across the polyQ region. In contrast, hydrogen-deuterium exchange shows absence of backbone amide protection, suggesting negligible persistence of hydrogen bonds. The seemingly conflicting results are explained by all-atom simulations, which show that Httex1 adopts tadpole-like structures with a globular head encompassing the N-terminal amphipathic and polyQ regions and the tail encompassing the C-terminal proline-rich region. The surface area of the globular domain increases monotonically with polyQ length. This stimulates sharp increases in gain-of-function interactions in cells for expanded polyQ, and one of these interactions is with the stress-granule protein Fus. Our results highlight plausible connections between Httex1 structure and routes to neurotoxicity.


Nature Structural & Molecular Biology | 2015

Polyalanine expansions drive a shift into α-helical clusters without amyloid-fibril formation.

Saskia Polling; Angelique R. Ormsby; Rebecca Wood; Kristie Lee; Cheryl Shoubridge; James N. Hughes; Paul Q. Thomas; Michael D. W. Griffin; Andrew F. Hill; Quill Bowden; Till Böcking; Danny M. Hatters

Collaboration


Dive into the Angelique R. Ormsby's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca Wood

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar

Archa H. Fox

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Ashish Sethi

University of Melbourne

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge