Medical Biology - Theses

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 130
  • Item
    No Preview Available
    Identification of molecular pathways associated with susceptibility and immunity to severe dengue and malaria
    Studniberg, Stephanie Irene ( 2023-12)
    In this era of increasing globalisation, urbanisation, and worsening climate change, the geographical range of transmission-competent mosquito vectors is shifting. Mosquito-borne diseases such as malaria and dengue are gradually emerging in previously unaffected areas, and re-emerging in areas where they had once previously subsided. With alarming increases in dengue case incidence, and for the first time, a chapter addressing the influence of climate change on malaria transmission in the World Malaria Report, it is clear that these two important vector-borne diseases are of utmost global relevance. As per the World Health Organization (WHO) guidelines, individuals presenting with warning signs signifying progression to severe dengue are required to remain under hospital observation. However, these warning signs appear late in the disease course and are non-specific. Consequentially, hospitals become overwhelmed with patients admitted for in-patient observation, many of whom do not progress to severe dengue. Biomarkers to detect progression to severe dengue upon hospital presentation are much needed to improve patient triage and resource allocation. In malaria, despite the great achievement of the recommendation by the WHO for the use of the RTS,S and R21/Matrix-M vaccines in children living in endemic areas, reductions in malaria case incidence remain at a prolonged stall. It is clear that efficacious vaccines approved for children to adults are required to reduce the global malaria burden. Further elucidation of the molecular mechanisms underlying the immune response to dengue and malaria is imperative if these outcomes are to be achieved. To address these outstanding concerns, an integrative systems immunology approach was utilised to identify molecular pathways associated with susceptibility and immunity to severe dengue and malaria. The studies within this thesis have integrated data from single-cell mass cytometry, serology, and transcriptional profiling of peripheral blood mononuclear cells from individuals progressing to either dengue fever (DF) or dengue haemorrhagic fever (DHF), as well as individuals living in a malaria-endemic regions of Indonesia with either symptomatic or asymptomatic Plasmodium falciparum and Plasmodium vivax malaria. Integrative data analysis identified frequencies and transcriptional profiles of effector CD4+ and CD8+ T cells as important components of dengue immunity in individuals progressing to DF. Furthermore, high frequencies of defined populations of CD4+ non-classical monocytes were associated with increased odds of developing DHF. Our approach discovered a strong transcriptional phenotype of immunosuppression underlying asymptomatic P. falciparum malaria, suggesting that the carriage of these infections could preclude complete parasite clearance. Lastly, unlike symptomatic P. falciparum malaria that induced a highly inflammatory response, clinical P. vivax infection featured the upregulation of anti-inflammatory pathways and checkpoint receptors, providing a feedback loop to ameliorate symptomatic infection. Furthermore, gene set enrichment analysis revealed profound dysfunction of the blood monocyte compartment in both symptomatic and asymptomatic P. vivax malaria. Together, the findings in this thesis have critical implications for the deployment and efficacy of malaria vaccines, and for the development of diagnostic tools to predict disease outcomes for dengue patients at point-of-care.
  • Item
    Thumbnail Image
    Manipulation of host defence mechanisms by Toxoplasma gondii
    Ruparel, Ushma ( 2023-10)
    Toxoplasma gondii is ubiquitous parasite of medical and agricultural importance, chronically infecting around a third of the global human population. Acute infection, caused by a form of the parasite known as a tachyzoite, in immunocompromised patients can cause blindness and encephalopathy. Treatments are limited to activity in the acute stage of infection, which is responsible for these disease processes. Chronic infection, characterised by differentiation of tachyzoites into bradyzoites, which are encysted parasites that reside in organs like the brain and the retina, is not only refractory to drug- but also immune-clearance. As a result, chronic infection is thought to persist for the lifetime of the host, serving as a reservoir for reactivation of acute infection during immune compromise. Being an obligate intracellular parasite, T. gondii must modify host signalling to create a parasite-permissive environment. This remodelling is, in part, attributed to the effector proteins exported by the parasite into the host cell. These proteins manipulate host signalling to evade the immune system and facilitate parasite persistence. In contrast to its role in acute infection, the impact of effector protein export in chronic infection remains largely uncharacterised. To better understand how effector protein export in chronic infection impacts host signalling, transgenic parasites were generated where the ability to export effector proteins was limited to either acute or chronic infection. Investigation of these parasites demonstrated that there is a defect in the ability of bradyzoites to survive host immune stress in the absence of effector protein export; restoration of this process confers a survival advantage in vitro. These findings also suggest that acute stage effector protein export is not sufficient in preventing clearance of bradyzoites, reiterating the idea that bradyzoites are not latently persisting in the host. Rather, they are active, dynamic entities that carefully regulate host signalling to prevent death of infected cells. In addition to its role in clearing bradyzoite infected cells, host programmed cell death (PCD) has also been implicated in acute T. gondii infection. Various PCD pathways have been investigated for their role in controlling acute infection, however, these findings are mostly in vitro and cell- or tissue-specific. It remains unclear how PCD affects parasite dissemination during acute infection in vivo, or whether there is redundancy in the PCD-mediated host defence against T. gondii, given the ability of the parasite to block certain arms of PCD using exported effector proteins. Mouse models of acute toxoplasmosis revealed a caspase-8 as a major mediator of resistance to acute infection, its role so critical that neither necroptotic nor pyroptotic death could act as a backup defence mechanism. This dissertation explores multiple avenues of host-pathogen interactions during acute and chronic T. gondii infection and provides new insights into how parasites establish successful chronic infection, as well as how the host attempts to control parasites before they can do so.
  • Item
    Thumbnail Image
    A conserved molecular mechanism of erythrocyte invasion by malaria parasites
    Seager, Benjamin Andrew ( 2023-10)
    Malaria is major global health burden causing over 240 million cases every year and leading to over 600,000 deaths mostly in pregnant women and children under the age of five. There is an urgent need to development novel therapeutic interventions for the control and elimination of malaria. During infection, malaria parasites must invade host erythrocytes in order to live within them. Invasion is a complex multi-step process that involves many molecular host-parasite interactions. In Plasmodium falciparum, the deadliest species of the parasite, the invasion protein PfRh5 assembles into a complex to bind its receptor basigin on the erythrocyte surface. Recent work has revealed two novel members of this complex, PfPTRAMP and PfCSS, form a heterodimeric platform for PfRipr, PfCyRPA, and PfRh5 binding. The PfPTRAMP-PfCSS-PfRipr-PfCyRPA-PfRh5 (PCRCR) complex, and its engagement with basigin, is essential for P. falciparum invasion. PfRh5 does not have an orthologue in all species of malaria, however PTRAMP, CSS and Ripr orthologues are present across the entire Plasmodium genus. This thesis sought to investigate these conserved proteins in other malaria species to further dissect the essentiality of these proteins for Plasmodium invasion more broadly. Orthologues of PTRAMP, CSS and Ripr from two important species of malaria, P. vivax and P. knowlesi, were investigated using recombinant expression and biophysical analysis. Assessment of complex formation shows a conserved assembly of the three proteins in both species, with similarities to P. falciparum. Structural determination of part of the complex revealed the basis of heterodimer formation between PTRAMP and CSS. Antibodies and nanobodies were produced and exhibit a high degree of cross-reactivity between species. A novel protein was identified that may bind to the complex and impart an erythrocyte binding function. The function of the complex and its components in invasion was confirmed using ex vivo invasion assays in Cambodian P. vivax field isolates. Taken together, this thesis shows that a three-membered complex consisting of PTRAMP, CSS and Ripr is conserved in three species of Plasmodium, likely forming a common invasion scaffold in all species of the genus, suggesting a conserved invasion mechanism with implications for cross-species vaccine development for the control of both P. vivax and P. falciparum malaria.
  • Item
    Thumbnail Image
    The role of programmed cell death and inflammation in the pathogenesis of SARS-CoV-2 disease
    Mendes Bader, Stefanie ( 2023-12)
    The impact of the SARS-CoV-2 pandemic was mitigated by an unprecedented scientific response that culminated in the implementation of vaccines and several other interventions. However, many questions remain which are relevant not only to SARS-CoV-2 but many other respiratory and pandemic viruses. Vaccines will continue to fall short of preventing infections as the virus evolves and antivirals cannot be distributed indiscriminately to mitigate mortality. There remains a pressing need to identify host factors that contribute to severe COVID-19 so patients can be stratified, and therapies delivered to those that would benefit the most. Severe COVID-19 is linked to a dysregulated hyperinflammatory immune response characterized by the release of pro-inflammatory cytokines. While cell death pathways have been postulated as central drivers of pathology, the molecular intricacies underlying these events remain elusive. To address this, we developed unique pre-clinical in vivo models that reproduce aspects of mild, severe and fatal COVID-19. Through serially passaging a clinical SARS-CoV-2 isolate in mice, we generated a mouse adapted strain that causes weight loss, inflammation and lung pathology in adult and is deadly in aged mice, reflecting key aspects of COVID-19. Our approach diverges from prevailing correlative or in vitro studies, offering a unique opportunity to delineate pathways causative of severe inflammation in vivo. Using gene targeted animals and transcriptomic analysis, we showed that the pro-inflammatory cytokines TNF and IL-1b drive severe disease. Interestingly, inflammasome pathways upstream of canonical IL-1b release do not influence disease outcomes, as loss of inflammasome components NLRP3 and ACS did not affect disease or viral burdens in vivo. Deletion of downstream targets of inflammasome signalling, such as pyroptosis initiators caspase-1/-11/-12 or effectors GasderminA/C/D/E did not ameliorate disease or viral burdens. Infection of animals lacking RIPK3 or MLKL showed that the lytic process of necroptosis, which lies downstream of TNF, did not contribute to disease. Collectively, these results suggested that lytic cell death did not contribute to SARS-CoV-2 disease, instead, the central determinant of severe disease outcome was caspase-8, a protein essential for the activation of apoptosis. Remarkably, instead of triggering cell death, infection drives caspase-8 to activate survival/inflammatory pathways and increase IL-1b levels. Combined deficiency of pyroptosis, necroptosis, and apoptosis mediators (caspase1/11/12/8/Ripk3-/-) provided no added benefit in ameliorating disease outcomes compared to caspase-8 deficient mice. In contrast, loss of caspase-1/-11/-12/Ripk3 caused a worsening of disease. Transcriptional profiling and comparison of lung tissues from compound mutant animals further confirmed that disease outcomes were not associated with differences in cell death pathways but rather with pro-inflammatory responses. The expression of full-length caspase-8 protein was upregulated in the lungs of SARS-CoV-2 infected mice and this was not accompanied by any increase in the fully cleaved, apoptotic form of caspase-8. The data collectively showed a pivotal role for caspase-8 in driving the pathogenesis of severe SARS-CoV-2 infection through the modulation of pro-inflammatory cytokines but not through the induction of apoptosis. This critical new understanding provides valuable insights for targeted therapeutic interventions and emphasizes the need for continued exploration of host-pathogen interactions in the context of COVID-19.
  • Item
    Thumbnail Image
    Understanding protein variants with high-throughput mutagenesis and machine learning
    Fu, Yunfan ( 2023-10)
    Genetic variations in protein-coding genes may cause amino acid substitutions in the matured proteins. These variants can potentially change the properties and functions of a protein. To evaluate the effects of these protein variants, multiple experimental and computational approaches have been utilised. Within these approaches, deep mutational scanning (DMS), a recently developed high-throughput mutagenesis method, enables the measurement of thousands of protein variant effects in a single experiment. To fully investigate the rich information in DMS results and have a better understanding of protein variant effects, here, I leveraged machine learning algorithms to build advanced computational models for DMS data. First, I reviewed that there are missing variant effect data in most DMS results, and I developed imputation models to fill in the missing values. I started by investigating the correlations between the variant effects measured within a DMS experiment and used these correlations to build imputation models. To understand the strengths and weaknesses of these models, I benchmarked them with previously published DMS imputation methods. At the end of this study, I built an ensemble imputation model by combining these novel and previously published methods to further improve the imputation accuracy. Many of the state-of-the-art variant effect predictors are built with DMS data, and I then managed to improve these predictors by further integrating variant effect data from alanine scanning (AS), a low-throughput mutagenesis approach. In this study, I established a rule-based classification tree to evaluate the compatibility between DMS and AS studies according to the similarity of their experimental assays. I showed that an improved variant effect predictor could be built only by modelling with high compatibility DMS and AS data. Finally, experimental measurements of protein variant effects may conflate protein stability and function. Here, I explored this relationship using DMS-measured variant effects and computed variant stability. I demonstrated that the correlation between variant effect and stability data differed on distinct protein regions and properties measured. Analysing these data with a dimensional reduction algorithm, I was able to automatically distinguish protein residues with different scales of fitness–stability association. Further investigation showed that this approach might be applied to discover protein functional sites and explain the mechanisms of loss-of-function variants.
  • Item
    No Preview Available
    Statistical models for pre-processing and simulating single cell RNA-seq data
    Wang, Jianan ( 2023-10)
    Since the first protocol published in the year of 2009, single-cell RNA sequencing (scRNA-seq) has become one of the most popular technologies in the omics world. scRNA-seq has been broadly applied in different areas, including understanding tumor microenvironment, inferring embryonic development, and discovering regulation pathways involved in plant seeding. The diverse applications facilitate the development of the scRNA-seq, and new protocols tend to sequence numerous of cells at low cost. With rapid growth of scRNA-seq technology, many issues arise and wait to be resolved when processing the data, such as removing batch effects, addressing dropout events, and annotation of cell populations. However, the requirements to tackle the above issues bring opportunities to apply statistical methods and devise new computational tools for analysing the data. In this thesis, we focus on pre-processing and simulation of single cell data. In the first part of the thesis, we mainly discuss different demultiplexing methods in the pre-processing step. The first method, CMDdemux, utilizes Mahalanobis distance to distinguish cells from different samples. This method performs well in low-quality data with hashtag contaminations during the cell hashing experiment. The second method is LCADemux, which uses latent class analysis (LCA) to combine cell hashing- and single nucleotide polymorphism (SNP)-based demultiplexing results. This hybrid framework is advantageous for analysing low input cell hashing data. The third method, LCAdoublet, applies the LCA method to combine doublet information from cell hashing and VDJ data. LCAdoublet is better at identifying inter-sample doublets and T-cell doublets than methods solely using transcriptomics data. In summary, our three pre-processing methods enable the accurate identification of doublets and assigning cells to their samples of origins, which contributes to cleaner data for downstream analysis. In the second part of the thesis, we talk about a novel single cell simulation method and its applications for data analysis. Our method is named GLMsim, and it applies a generalized linear model to simulate the batch and biological effects simultaneously. Compared to other methods, our method is able to simulate single cell data resembling the original data, especially for the data collected under complex conditions. Our single cell simulators have multiple applications, such as benchmarking single cell integration methods, providing guidance for differential expression analysis and checking the assumptions of models. In short, our simulation method should help researchers develop better tools for downstream analysis. Overall, we applied multiple statistical methods to fill gaps existing in dealing with single cell RNA-seq data. Our methods have been demonstrated to overcome challenges when analysing single cell RNA-seq data. We hope that they can be applied in the medical field to extend our knowledge of human health and disease.
  • Item
    Thumbnail Image
    Structural and Pharmacological Investigation of the JAK/SOCS1 Interaction
    Wu, Yuntong ( 2023-09)
    Cytokines are important signalling molecules which can cause inflammation, regulate haematopoiesis and modulate the immune response. Over 50 cytokines achieve their biological functions on cells via the JAK/STAT pathway. SOCS (Suppressors of Cytokine Signalling) family are induced by the cytokine-JAK-STAT cascade and act as negative feedback regulators to specific cytokine pathway. The existence of the SOCS family prevents cells from persistent activation of cytokine signalling but can also play a role in cytokine resistance and tumour progression. SOCS1, a member of SOCS family, is specifically induced by IFN-gamma. And the SH2 domain and the KIR domain are critical for SOCS1’s function. The SH2 domain is responsible for protein-protein interactions by specifically targeting pTyr-containing substrates. However, the structural details of the SOCS1 SH2 domain interacting with its targets and how this interaction happens in vivo remained unclear. Our work determined the crystal structures of SOCS1 SH2 domain bound to various ligands, which uncovered the molecular interactions within this domain. Additionally, small molecules with potentially high affinity for targeting the SOCS1 SH2 domain were identified through structure-guided drug design. The KIR domain of SOCS1 can insert into the substrate-binding pocket and directly inhibit the catalytic activity of JAK1. Our research also attempted to develop novel type of JAK inhibitors based on the binding mechanism of KIR domain. These findings provided insights into the interactions between JAK and SOCS1 and also highlighted the potential of developing small molecules to disrupt these interactions.
  • Item
    Thumbnail Image
    Structural Studies of Type-I Haematopoietic cytokine receptors
    Sarson-Lawrence, Kaiseal Tane Garvey ( 2023-09)
    Haematopoiesis is a complex process by which the full complement of mature blood cells is produced from a small population of hematopoietic stem cells in the bone marrow. Cytokine signalling plays a crucial role in haematopoiesis and at least 14 different cytokines are involved in determining the fate of hematopoietic stem cells. Cytokines act on cells by binding and oligomerising cytokine receptors on the cell surface. This oligomerisation activates intracellular JAK kinases that kick off a signalling cascade resulting in a cellular response. As cytokine binding is the critical first step in receptor activation, understanding how different cytokines bind to their receptors and how this extracellular binding event translates into an intracellular signal is fundamental to understanding hematopoietic diseases and designing therapeutic molecules. Thrombopoietin (TPO) and granulocyte colony stimulating factor (GCSF) are haematopoietic cytokines that regulate the production of platelets and neutrophils, respectively, from their precursor cells. The thrombopoietin receptor (TPOR) belongs to the short family of cytokine receptors and contains a unique duplication of its ligand binding domain not usually seen in other cytokine receptors. While the structure of TPO has previously been determined by X-ray crystallography, the structure of the receptor and receptor-cytokine complex have not. The GCSF receptor (GCSFR) belongs to the “tall” family of cytokine receptors. This family is characterised by three additional fibronectin type-III (FnIII) domains in the extracellular domain, which extends the receptor out from the cell membrane. Although a partial structure of the GCSFR-GCSF complex has been determined previously, this structure lacked the three membrane-proximal FnIII domains. I expressed and purified recombinant extracellular domains of both GCSFR and TPOR along with their respective cytokines. Cryogenic electron microscopy (cryo-EM) was then used to solve the structures of both receptor-ligand complexes and study the mechanism of cytokine binding and receptor activation of the two receptors. This research has resulted in the first experimentally determined structures of the entire extracellular domains of both the TPOR and GCSFR receptors in complex with their cytokines. These structures and the corresponding biophysical data have improved our understanding of how these two receptors are activated by their cytokines.
  • Item
    Thumbnail Image
    Understanding epilepsy genetic risk: integrating common and rare genetic variation
    Oliver, Karen Louise ( 2023-10)
    The epilepsies are a group of neurological disorders affecting up to 4% of people during their lifetime. There are many different epilepsy syndromes. At the broadest level, syndromes can be classified into those that are focal, where there are focal-onset seizures, or generalised, where there are generalised-onset seizures; these represent the most common epilepsies. The developmental and/or epileptic encephalopathies (DEEs) are the most severe group of epilepsies where seizure activity is associated with developmental slowing or regression. The DEEs typically begin in infancy and childhood and have many rare single-gene causes. With both rare and common variants established contributors to epilepsy genetic risk, the over-arching aim of this thesis was to explore the potential interaction of these different variant types. First, I curated all reported single-gene monogenic causes. The clinically heterogeneous DEEs had >800 reported single-gene causes. In contrast, <50 single-gene causes for focal and generalised epilepsies are known. Clinical genetic studies suggest these epilepsy types are polygenic, with current evidence suggesting many contributions from both rare and common genetic variants. Common variants are detectable by genome-wide association studies (GWASs). I was Consortium Coordinator and one of the analysts for the third International League Against Epilepsy GWAS involving >29,000 patients with epilepsy. Here, I led an analysis to prioritise genes near GWAS signals that demonstrated a convergence between common and rare gene pathways, and a study of cross-trait genetic correlations that highlighted the pleiotropic nature of many epilepsy-associated loci. Next, to explore potential rare and common genetic variant interplay, I focused on families with epilepsy. This familial approach was motivated by the observation that, even when sharing the same familial rare pathogenic variant of major effect, the clinical presentation for relatives can be highly variable. This supports a role for, yet unidentified, epilepsy genetic modifiers. In other complex traits, like cancer and heart disease, studies have shown that common polygenic background, as captured by polygenic risk scores (PRSs), derived from large GWASs, can modify the penetrance of rare monogenic causes. To determine if common polygenic background plays a modifying role in the epilepsies, I next demonstrated that epilepsy PRSs are enriched in patients with a positive family history for epilepsy compared to those without. Whilst families with epilepsy have previously been targeted for rare variant discoveries, we provide the first support for common genetic variation playing a role in the familial aggregation of epilepsy. Furthermore, common risk variants for focal epilepsy were shown to be enriched in a specific familial focal epilepsy syndrome, despite no variants in the largest focal epilepsy GWAS reaching genome-wide significance. Finally, I explored whether the role played by common epilepsy risk variants is disease modifying. This was done by studying 58 families with the clinically heterogenous syndrome of genetic epilepsy with febrile seizures plus (GEFS+), many with a known rare variant of major effect. In these families, I showed that higher epilepsy PRSs correlated with more severe epilepsy phenotypes. This provides the first support for common genetic background modifying the clinical expression of rare pathogenic variants in the epilepsies.
  • Item
    Thumbnail Image
    3D Imaging and Cellular Barcoding: Novel Tools for Exploring Cancer Heterogeneity
    Lewis, Sabrina Milly ( 2023-09)
    Breast cancer affects 1 in 7 Australian women, and the risk of death from metastatic (stage 4) disease remains high. Progression to advanced disease is difficult to treat, especially when the availability of targeted treatments is limited for some cancer subtypes. Metastases form when cancer cells shed from the primary tumour, enter the blood and lymphatic vessels, exit and proliferate in distant organs. Understanding the interactions between these heterogeneous lesions and the vessels that facilitate their spread, will enable a better understanding of this process and potentially lead to improved cancer treatments. Not all tumour cells have the same ability to generate metastases. Specific clones (defined as cancer cells that have derived from the same ancestral cell) differ in terms of the organs they target, their behaviour in specific microenvironments, and how they cooperate with other clones. To date, the methods used to study the clonality and heterogeneity of cancer metastases often involve tissue dissociation or 2D imaging. Consequently, the spatial resolution of clones in their native microenvironment is lost. New methodologies and technologies are required to facilitate spatial discoveries, to advance our understanding of cancer heterogeneity, metastasis, and the tumour microenvironment. Here, I developed a novel pipeline for three-dimensional whole organ imaging of human-in-mouse models of metastatic breast cancer. Light-sheet microscopy was used to capture large volumetric datasets, reducing the information loss observed in 2D tissue sections. I used lentiviral gene ontology (LeGO) vectors, an optical barcoding method, to identify seven individual clones. In combination with vessel casting (a perfusion-based method that enables vasculature imaging), tissue clearing, and an analysis pipeline, I reveal the relationship of aggressive breast cancer clones and the blood vasculature in murine lungs and brain. This represents a method with unprecedented detail and clonal resolution at large volume scales. My results indicate that large vessels may be correlated with enhanced metastatic growth. I also show that metastases that wrap around blood vessels are more likely to be polyclonal (containing multiple clonal populations), which are more aggressive than monoclonal (single clone) metastases, with potential implications for treatment targets. Underlying the clones’ specific behaviours, are differences in gene expression. Based on these results, I propose that transcriptional information, in combination with clonal identity and spatial tissue context, is required to reveal the molecular pathways that are responsible for these clonal behaviours, which may represent novel therapeutic targets. To simultaneously track a higher number of cancer clones (i.e. thousands) and their gene expression in situ, I developed a novel smFISH and RNA barcoding method (FISHcodes). I show that the detection of hundreds of transcripts alongside dozens of clones (scalable to thousands) is feasible and will enable novel insights regarding clonal behaviour in cancer biology. Together, the results presented throughout this thesis have demonstrated that novel methodologies enabling the study of cancer cell heterogeneity and its interplay with the microenvironment, can address new questions about in situ cancer clone metastasis and growth.