Medical Biology - Theses

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 30
  • Item
    Thumbnail Image
    Functional and structural characterisation of VDAC2 in BAK-mediated apoptosis
    Yuan, Zheng ( 2023-06)
    BAK and BAX are the executors of intrinsic apoptosis. Their activity is tightly regulated by their interactions with the BCL-2 family proteins, but also non-BCL 2 proteins including the mitochondrial Voltage-Dependent Anion Channel 2 (VDAC2). Whilst targeting their interactions with BCL-2 family proteins to manipulate apoptosis clinically to treat diseases including cancer and potentially degenerative diseases is receiving attention, their interaction with VDAC2 remains unexplored. VDAC2 is important for the targeting of both BAK and BAX to mitochondria where they execute their apoptotic function, and its interaction with BAK has recently emerged as a therapeutic target to manipulate BAK-mediated apoptosis. The Chapter 3 presents the intracellular evidence of how VDAC2 interacts with BAK to modulate BAK-driven apoptosis. I have identified key residues involved in the interaction between BAK and a cytosol-exposed region on VDAC2 using mutagenesis and obstructive cysteine labelling. Stabilisation of this interaction through mutagenesis of VDAC2 not only restrains BAK activity but is also sufficient to inhibit a cells response to BH3 mimetic compounds. Cysteine crosslinking experiments reveal that VDAC2 binds to BAK hydrophobic groove, which is to date the first example of a non alpha-helix binding to the BAK groove. The Chapter 4 details attempt to investigate the interaction between BAK and VDAC2 using recombinant proteins. Furthermore, given that BAK and VDAC2 are known to engage other proteins in the MOM, including VDACs 1 and 3, the Chapter 5 describes sample preparation and cryo-EM analysis of the complex isolated from mitochondria from mammalian cells to attempt to resolve the structure of this multi-protein complex. Here I report the preliminary cryo-EM data of the purified BAK–VDAC2 multi-complex. This thesis provides new insights into how BAK is regulated through its interaction with VDAC2 through both biochemical and structural perspectives, and can guide new avenues for potential therapeutic intervention.
  • Item
    Thumbnail Image
    A genetics-based investigation into the regulation of RIPK1 and caspase-8 during cell death and disease
    Simpson, Daniel Steve ( 2022)
    Cell death is a fundamental process needed for healthy development, immunity and life. The tight control and regulation of cell death signalling is important for cellular homeostasis, and the de-regulation of cell death is a hallmark of many diseases ranging from infection to cancer. Several regulated cell death (RCD) pathways have been described, with genetically encoded cell death signalling molecules and effectors dictating cellular fate. Some of these, such as necroptosis and pyroptosis, are highly inflammatory and immunomodulatory, while others, such as apoptosis, are generally considered non-inflammatory and tolerogenic. Caspase-8 is a critical cell death protein that also has a pleiotropic role in inflammation. Receptor interacting protein kinase (RIPK)1 liaises with external signals to control the death and inflammatory functions of caspase-8, but major gaps remain in our understanding of how RIPK1 regulates the death and non-death functions of caspase-8. Identifying and characterising the mechanisms that control caspase-8 activity is crucial to understanding how we might best therapeutically target cell death signalling to overcome relevant diseases. This PhD thesis explores the regulation of caspase-8 activity and identifies key upstream checkpoints to therapeutically intersect and modulate caspase-8 activity. Firstly, this thesis genetically delineates a unique caspase-8-dependent cell death triggered by combined signalling of host-derived interferon (IFN)-gamma and pathogen ligands that engage Toll-like receptors (TLRs). Experiments show that caspase-8 cell death signalling is licensed by nitric oxide (NO), which is produced by the inducible nitric oxide synthase (iNOS) protein. Physiologically, both caspase-8 and iNOS contributed to disease severity in a model of severe acute respiratory syndrome-associated coronavirus-2 (SARS-CoV-2) infection, suggesting iNOS might licence damaging cell death and inflammation during coronavirus disease of 2019 (COVID-19). Secondly, the physiological role of Mind Bomb-2 (MIB2), a recently described pro-survival protein that prevents caspase-8 activation by RIPK1 in cancer cells, is examined using novel MIB2 gene targeted mice. This thesis reveals the physiological function of MIB2 in vivo and examines the function of MIB2 in both inflammation and cancer disease models to determine whether therapeutics designed to inhibit MIB2 could be used to safely activate caspase-8. These studies find that deficiency or inactivation of MIB2 is well-tolerated in mice and does not impact important biological processes including development, haematopoiesis, viability or fertility. Interestingly, challenging MIB2 knockout mice to drive excessive caspase-8 activity leads to enhanced cell death-induced dermatitis, while inactivation of MIB2 limits tumourigenesis in a model of inflammation-driven colorectal cancer. This thesis provides critical insight into the regulation of caspase-8 and uncovers distinct modes of regulation detailing how elevated NO or, inhibition of MIB2 contribute to excessive cell death and disease. This work aids the design of next generation treatments to overcome cell death resistance and transforms our understanding of how caspase-8 is regulated in inflammation and disease.
  • Item
    Thumbnail Image
    The role of RIPK3 ubiquitylation and MLKL signalling during cell death and autophagy
    Frank, Daniel ( 2021)
    Receptor Interacting Serine/Threonine Kinase-3 (RIPK3) is essential for necroptosis, an inflammatory form of programmed cell death pathway implicated in innate immunity, kidney ischemia reperfusion injury, and systemic inflammatory response syndrome. In the classical model, cells committed to necroptosis phosphorylate RIPK1, which in turn drives RIPK3 phosphorylation and oligomerisation. Active RIPK3 oligomers subsequently phosphorylate mixed lineage kinase domain-like protein (MLKL) pseudokinase which induces its translocation to the plasma membrane. The necroptosis pathway culminates in MLKL perforating the plasma membrane as a prelude to cellular rupture and release of inflammatory cytokines and damage-associated molecular patterns to the extracellular milieu. In addition to being a pro-necroptotic kinase, RIPK3 is also capable of triggering apoptosis when its kinase activity is restrained. Moreover, numerous death-independent roles of RIPK3 have been described in the context of inflammation such as arthritis, viral infection, or colitis whereby RIPK3 either promotes or dampens the secretion of pro-inflammatory cytokines. Understanding the molecular regulation of RIPK3 will thereby facilitate the ongoing pre-clinical development of RIPK3 inhibitors. Like most proteins, post-translational modification (PTM) is a critical fine tuner of RIPK3 activities. Ubiquitylation, in particular, has recently garnered attention in the cell death field as loss of this PTM may result in hyperactive RIPK3 which consequently accelerates death and inflammation. However, the post-translational control of RIPK3 signalling is not fully understood. Using mass-spectrometry, I identified a novel ubiquitylation site on murine RIPK3 on lysine 469 (K469). Complementation of RIPK3-deficient cells with a RIPK3-K469R mutant demonstrated that the decoration of RIPK3 K469 by ubiquitin limits both RIPK3-mediated caspase-8 activation and apoptotic killing, in addition to RIPK3 autophosphorylation and MLKL-mediated necroptosis. Unexpectedly, the overall ubiquitylation of mutant RIPK3-K469R was enhanced, which largely resulted from additional RIPK3 ubiquitylation upstream on lysine 359 (K359). Loss of RIPK3-K359 ubiquitylation reduced RIPK3-K469R hyper-ubiquitylation and also RIPK3-K469R killing. Collectively, I therefore propose that ubiquitylation of RIPK3 on K469 functions to prevent RIPK3 hyper-ubiquitylation on alternate lysine residues, which otherwise promote RIPK3 oligomerisation and consequent cell death signalling. I further investigated the consequence of abolishing RIPK3 K469 ubiquitylation by generating Ripk3K469R/K469R mice. In agreement with in vitro findings, primary fibroblasts with mutant RIPK3-K469R enhanced apoptosis, and in vivo studies demonstrate that RIPK3-K469 ubiquitylation contributes to pathogen clearance. Specifically, when Ripk3K469R/K469R mice were challenged with Salmonella enterica serovar Typhimurium, bacterial loads in the spleen and liver were significantly increased relative to wildtype control animals. The increased bacterial burden in the mutant mice was consistent with reduced IFNg produced in the serum, while the elevated MCP-1 cytokine upon infection might be indicative of heightened immune infiltrates. Although necroptosis signalling clearly triggers cell death, how it might impact other cellular responses remains unclear. Therefore, to further delineate the functional outcomes of necroptotic activity I examined how its signalling impacts autophagy. The autophagy pathway is triggered when cells are deprived of nutrients. Although regarded as a pro-survival pathway which acts to recycle and remove damaged organelles, studies have recognised that autophagic pathways can impact cell death processes. In apoptosis, for instance, autophagy acts to limit pro-inflammatory IFN-b secretion, thus decreasing apoptotic immunogenicity. Nonetheless, little is known about the status of autophagy during necroptosis. I demonstrate through various genetic, imaging, and pharmacological approaches that active MLKL translocates to autophagic membranes during necroptosis. However, contrary to previous findings which reported the activation of autophagy upon necroptotic activity based on increased lipidated LC3B, a commonly used marker of autophagy induction, I challenged this conclusion by demonstrating that the accumulation of active LC3B during necroptosis is a consequence of reduced autophagic flux. Therefore, unlike apoptosis which proceeds in tandem with autophagy, the induction of necroptosis negates autophagy in an MLKL-dependent manner. While the function of MLKL-mediated autophagy inhibition warrants further investigation, I propose that attenuating autophagy during necroptosis contributes to the immunogenicity of this cell death modality by limiting the ability of the cell to clear damaged organelles and immunogenic molecules. Overall, my research has helped in outlining how a key necroptotic molecule RIPK3 is regulated post-translationally and how this is relevant in the context of microbial defence. I have also defined novel functional roles for necroptosis signalling in the regulation of autophagic responses. Understanding the molecular regulation of necroptosis signalling and how this cell death pathway is linked to other cellular responses, such as autophagy, is important for the accurate design of new therapeutics to target these pathways in pathological settings.
  • Item
    Thumbnail Image
    Control of the Intrinsic Pathway of Apoptosis
    Djajawi, Tirta ( 2019)
    Apoptosis is a cellular process of programmed cell death. The intrinsic pathway of apoptosis is triggered by mitochondrial outer membrane permeabilization, a point of no return that coincides with the release of cytochrome c into the cytosol where it activates the main effectors of cellular destruction: the caspases. The mitochondrial pathway that is centered on MOMP is tightly regulated by BCL2 family proteins, which includes some members that promote apoptosis and others that inhibit it. The interplay between these proteins with opposing roles determines whether a cell will die or survive. In a healthy cell, pro-survival BCL2 proteins inhibit the effector proteins BAX and BAK. BH3-only proteins are activated in response to cellular stress and promote apoptosis by neutralizing pro-survival proteins. Targeting BCL2 proteins to provoke apoptotic cell death has proven to be a successful strategy for cancer therapy with the BCL2-selective drug venetoclax exhibiting remarkable efficacy in treating cancers that rely on BCL2 for their survival. MCL1, a protein related to BCL2, is likewise critical for the survival of many cancer cells, making it another attractive anti-cancer drug target. Selective MCL1 inhibitors have been developed and are currently being evaluated in clinical trials to establish their safety and efficacy. Safety is a particular concern for MCL1 inhibitors because MCL1 is also essential for the survival of many cells in critical organs and tissues throughout the body. It remains to be seen if a sufficient therapeutic window will exist when MCL1 is targeted systemically. An alternative and potentially safer strategy to modulate MCL1 survival function would be to target pathways that regulate its activity in particular contexts. In Chapter 3 and 4, I focus on one such mechanism of MCL1 regulation: its turnover by the ubiquitin proteasome system. My work in Chapter 3 elucidated details of how MCL1 protein turnover is regulated by BH3-only protein NOXA. Using CRISPR-Cas9 screen, I discovered that the mitochondrial E3 ligase MARCH5, the E2 conjugating enzyme UBE2K and the mitochondrial outer membrane protein MTCH2 co-operate to mark MCL1 for degradation by the proteasome. I also demonstrated that this pathway is constitutively active in cells where NOXA is abundantly expressed and showed that manipulating NOXA expression in those cells impacts on MCL1 survival function. Having successfully demonstrated the power of CRISPR-Cas9 screen in Chapter 3, I undertook further screens in Chapter 4 to identify proteins, such as deubiquininating enzymes (DUBs), that might serve to enhance MCL1 protein stability. I did not identify any strong hits from these screens, possibly because multiple DUBs act redundantly on MCL1. Consistent with this hypothesis, only mild impacts on MCL1 protein stability were observed upon deleting DUBs previously reported to act on MCL1. Finally, in Chapter 5, I investigated how BH3 mimetics mimic the activity of BH3-only proteins to induce apoptosis. I studied how selective BH3 mimetic compounds perturb interactions throughout the BCL2 protein network beyond their direct protein targets. I showed that these second order impacts are crucial for effective killing. Apoptosis induced by the BCL2 selective inhibitor venetoclax, for example, typically also involves inhibition of MCL1. The impact on MCL1 in this context occurs as a consequence of displacing BH3-only proteins normally bound to BCL2.
  • Item
    Thumbnail Image
    Advancing BH3 mimetics to treat cancers
    Luo, Mingjie ( 2019)
    The evasion of apoptosis, one of the hallmarks of cancer, is observed in many cancers. This can also impair the efficacy of many conventional chemotherapies. The BCL2 protein family is the central regulator of the intrinsic apoptotic pathway and plays a vital role during tumor development. In particular, the levels of the pro survival family members are often elevated in some cancers. Venetoclax, a BH3 mimetic inhibitor that mimics the BH3-only proteins, natural inhibitors of the pro-survival BCL2 proteins, has proven to be effective for treating hematological cancers by selectively targeting BCL2. This has translated into regulatory approvals of venetoclax for treating a subset of chronic lymphocytic leukemia and acute myeloid leukemia. In addition to targeting BCL2, potent and specific BH3 mimetic inhibitors of its relatives, BCLxL and MCL1, are now also available. However, their full clinical utility is poorly defined. This thesis focuses on advancing the utility of the BH3 mimetic compounds as anti-cancer agents. Previous studies have suggested roles for BCLxL and MCL1 in many solid cancers (e.g. colon, breast, lung). In particular, colorectal cancers have elevated levels of the pro survival protein, one usually associated with chemo resistance. Furthermore, colorectal cancer patients with advanced disease or those who carry poor prognostic markers do not respond well to the current stand of care therapies such as surgery and adjuvant chemo/radiotherapy. Given the pressing need to find better treatments for these patients, we first utilized a panel of validated BH3 mimetics to assess the feasibility of using them for treating colorectal cancer. By using cancer cell lines and patient-derived organoids, we identified and validated BCLxL and MCL1 as the most important survival factors for colorectal cancer. We then validated them as potential targets by pharmacological inhibition in a mouse model in vivo. Moreover, we found that even those tumors that harbor poor prognostic factors respond as avidly as those do not, further highlighting the potential of this approach for treating patients with colorectal cancer. Even though the targeting BCLxL might be a possible approach to kill cancers that depend on it, the clinical use of BCLxL selective inhibitors is limited due to the toxicity of BCLxL inhibition on platelets. I have screened for novel regulators of BCLxL using the CRISPR/Cas9 technology, which might offer potential approaches to target BCLxL safely. The final goal of this thesis is to identify biomarkers that predict response to BH3 mimetics, given that there are few reliable tools to stratify patients that might respond well to these novel anti-cancer agents. By using large scale transcriptomic datasets from publicly available RNA sequencing studies, I was able to identify a few candidate genes and achieve reasonable prediction performance.
  • Item
    Thumbnail Image
    Mechanisms of angiogenesis in development and disease
    Grant, Zoe Louise ( 2019)
    The expansion of blood vessel networks by angiogenesis is critical for matching blood supply to the metabolic demands of growing tissues. In adult tissues, blood vessels and the endothelial cells (ECs) that make them generally remain in a quiescent state. However, the reactivation of angiogenesis is a hallmark of a number of diseases including cancer and vision-threatening eye diseases. In the case of eye disease, angiogenesis results in abnormal vascular growth that damages the retina. This abnormal angiogenesis often occurs in response to retinal hypoxia that can arise as a result of excessive retinal vessel regression. Understanding the mechanisms by which blood vessels both grow and regress is therefore important for understanding how vascular diseases arise in the eye. In this thesis, I have used in vivo mouse models of normal and pathological angiogenesis in the retina to examine the molecular and cellular mechanisms that control blood vessel growth and regression. In the first part of my thesis, I investigated the role of the histone acetyltransferase HBO1 in retinal blood vessel growth. Through its histone acetyltransferase activity, HBO1 promotes transcriptionally permissive chromatin and is necessary for cells to change their gene expression patterns, particularly during development. I found that HBO1 was required for the expression of genes and pathways that are upregulated by ECs undergoing angiogenesis. ECs lacking HBO1 failed to undergo directed migration during angiogenesis, resulting in reduced blood vessel production both in the normal retina and in a model of pathological retinal vessel growth. In the second part of this thesis, I examined the role of EC apoptosis in disease-causing blood vessel regression using a model of ischaemic retinopathy. In this model, vessel regression associated with EC apoptosis results in retinal ischaemia, causing a hypoxic response that drives abnormal vessel growth similar to that seen in human eye diseases. I found that blocking EC apoptosis did not prevent vessel regression or the onset of retinal ischaemia. Nonetheless, it completely prevented the loss of ECs from ischaemic areas of the retina. These preserved ECs were capable of rapidly reassembling into a functional vessel network that reduced hypoxia and the subsequent pathological vascular response. Vessel reassembly was not impeded by neutralising VEGFA, suggesting that it is not dependent on high levels of VEGFA produced by the ischaemic retina. Overall, my studies provide new insight into the mechanisms of blood vessel growth and regression and may potentially open new therapeutic avenues for preventing abnormal blood vessel growth in retinal vascular disease.
  • Item
    Thumbnail Image
    Defining programs of cell death that can be harnessed to impact on outcomes of chronic viral infection
    Preston, Simon Peter ( 2019)
    Pathogens causing chronic infections have successfully evolved mechanisms to subvert host immunity. Excessive and inappropriate inflammation together with attrition of repeatedly overstimulated high affinity T cells leads to abrogated immunity and persistence of pathogens such as HIV and HBV. T cell exhaustion has been touted as a prelude to T cell deletion during these infections, however, studies indicate that high affinity T cell clones are deleted at the onset of infection. The T cells that remain have lower affinity for pathogen epitopes and hence their response is weaker and more easily antagonised by inhibitory networks, including T-regulatory (Treg) cells. The killing of immune effector cells during chronic overwhelming infections is juxtaposed to the pathogen’s attempts to promote survival of infected target cells. Keeping infected cells alive is imperative for the maintenance of a microbial replicative niche. In this body of work, I dissected the role of host cell molecules and how they contribute to the death and survival of immune and infected cells. Necroptosis did not contribute to the loss of highly functional virus-specific CD8+ T cells during the course of infection. In contrast, when I interfered with death receptor signalling there was a modest rescue of functional CD8+ T cells. This gain in immune function, however, did not translate to improved viral control. The same mechanism I used to promote the survival of T cells made infected target cells refractory to death receptor mediated killing and therefore, offset any gain in immune function. Whilst examining the role of necroptosis in chronic infection, I made the discovery that the necroptotic inducer molecule, RIPK3, has additional non-necroptotic roles. Ripk3-/- mice cleared LCMV with enhanced kinetics compared to wild-type mice and mice that lacked the necroptotic executioner MLKL. I found that in the absence of RIPK3, chronically infected mice had impaired IFNβ responses. Excessive and prolonged IFNβ production is known to impair immunity. This may partially explain why mice lacking RIPK3 had enhanced numbers of granzyme B expressing T cells and controlled infection better than WT animals. The host-viral dynamics that favour displacement of highly functional cells with poorly activated cells makes the immune system highly vulnerable to inhibition through the activity of Treg cells. I next investigated the role of Treg cells in immune dysfunction during chronic infections and I was particularly interested in the cell death and cell survival pathways that contributed to the turnover and accumulation of these cells. I utilised mice with a Treg-specific deletion of Casp8. These mice had twice as many Treg cells as wild-type mice at steady state. Surprisingly, when these mice were infected with chronic LCMV, only 25% of the animals survived to 145 days post infection. Moribund animals succumbed to overt T cell activation and autoimmunity due to a precipitous drop in Treg cell numbers. Survivors, intriguingly, eliminated LCMV in most organs consistent with a massive gain in immune function. The death of the Treg cells was due to necroptosis. When I ablated the necroptotic pathway, through the deletion of Mlkl, I completely prevented the loss of Treg cells and the fatal immune pathology in Treg conditional caspase-8 deficient mice. I found that differential expression of RIPK3 and MLKL in Treg cells made them highly susceptible to necroptosis during chronic infection compared to Tconv cells. This was also the case for human Tregs and I was able to preferentially kill these cells, over Tconv cells, in vitro by driving necroptosis with a clinical stage caspase-8 antagonist called emricasan. Necroptosis is a lytic form of cell death that promotes inflammation and it has been implicated in chronic liver disease. I initially investigated if necroptosis in the liver contributed to the control of chronic LCMV, HBV or the malaria parasite Plasmodium berghei. Ablation of necroptosis had no impact on liver-pathogen dynamics and no impact on general liver function and architecture. In many cell types caspase-8 inhibits death receptor induced necroptosis. So, I reasoned that this molecule must be inhibiting induction of necroptosis in the liver of infected animals. I examined this by infecting mice that had a conditional loss of caspase-8 within hepatocytes. Despite abundant, infection driven, death ligands I observed no necroptosis in the liver. Even drug induced ablation of NF-ĸb survival signalling, downstream of TNF, failed to promote liver necroptosis in the aforementioned scenarios. The liver’s inability to undergo necroptosis was confirmed in mice with a human chimeric liver. I showed this refractoriness was due to liver repression of RIPK3 in humans and mice. The work conducted in this thesis provides important insights into the cell death pathways that are engaged in diverse cell types during chronic viral infections and I provide evidence that antagonising them therapeutically may lead to better clinical outcomes.
  • Item
    Thumbnail Image
    Molecular mechanisms of thymic tolerance induction and recovery from involution
    Heinlein, Melanie ( 2019)
    Thymic epithelial cells (TECs) are vital for the formation of the thymic microenvironment and the differentiation of T cells. This process involves a series of interactions between TECs and thymocytes that govern T cell commitment, progenitor T cell proliferation and differentiation and TCR specificity-based selection of immature T cells to finally allow the release of mature T cells capable of mounting an immune response against foreign antigens (e.g. from pathogens), whilst remaining tolerant of self. Although the role of TECs in mediating these processes is well established, major gaps in our understanding of the molecular mechanisms involved in TEC development, maintenance and function remain. In this thesis we seek to address this knowledge gap and define: (a) the molecular mechanisms underlying TEC survival and death during injury; and (b) the molecular mechanisms that control the expression of peripheral tissue self-antigens (PTAs) in TEC which is required to mediate self-tolerance. Atrophy of the thymus following irradiation or high-dose chemotherapy impairs immune recovery in patients, which is a significant cause of morbidity and mortality. Despite the importance of TECs for immunity, there is little understanding of the mechanisms that control TEC survival and death in the context of treatments that damage the immune system. By using different genetically modified mouse models targeting the intrinsic pathway of apoptosis in both TECs and thymocytes, we found that: (a) TECs die via the intrinsic apoptotic pathway after irradiation; (b) the pro-survival proteins BCL-2 and BCL-XL are crucial for TEC regeneration; and (c) blocking thymocyte death can partially rescue TECs following irradiation. These data provide new insights into the molecular control of TEC survival and regeneration that could be used to inform new treatments that maintain or restore thymic function in immunosuppressed patients. A unique property of TECs is their capacity to express thousands of PTAs to mediate immune tolerance of non-lymphoid self-antigens. The autoimmune regulator, AIRE, is required for the transcription of the majority of these PTAs, and defects in AIRE’s function lead to autoimmune disease in mice and humans. The precise molecular mechanisms by which AIRE orchestrates such broad, tolerogenic transcription of PTAs in TECs remain only partially understood. We here show that the acetyltransferase, KAT7, which mediates the majority of histone 3 lysine 14 acetylation (H3K14ac), is essential for AIRE-mediated expression of PTAs, the establishment of a normal thymic microenvironment and self-tolerance. This study reveals an important role for histone acetylation in AIRE’s function in TEC and immunological tolerance. In conclusion, these studies highlight essential molecular mechanisms underlying TEC survival, regeneration and function that are crucial for immune function and tolerance. This newly gained knowledge will further aid the design of treatment strategies for immune recovery after cytoablative and cancer treatment as well as autoimmune diseases.
  • Item
    Thumbnail Image
    The role of BCL2 family proteins in apoptosis regulation during angiogenesis
    Watson, Emma Caroline ( 2016)
    Blood vessels are multicellular tubes, lined with endothelial cells (ECs), that form a hierarchical network essential for the distribution of blood, oxygen, nutrients, hormones and immune cells around the body and removal of metabolic waste products from tissues. Angiogenesis, the growth of new vessels from pre-existing ones, is essential to match the size of the blood vessel network to the metabolic demands of growing tissues. During this process, an over-production of vessels results in the formation of a dense vessel plexus that is inefficient for blood flow. From this dense network, excess vessels undergo a process of selective regression termed ‘pruning’ to produce a mature, hierarchical vessel network. EC apoptosis occurs as part of the angiogenic remodelling processes, but its contribution to angiogenic vessel remodelling, be it vessel pruning or some other purpose, has remained unclear. In this thesis I directly investigated the role of EC apoptosis during angiogenesis by analysing mice in which ECs were unable to execute the apoptotic program regulated by BCL2 family proteins. I found that while EC apoptosis improved the efficiency of selective vessel pruning, it was ultimately dispensable for this process. Instead, blood vessels formed in the absence of EC apoptosis contained excessive numbers of ECs resulting in increased diameter of mature capillaries. Having established that the BCL2 family was essential for promoting EC death during angiogenesis, I investigated whether pro-survival members of the family were required for the survival of ECs during angiogenesis. Using the neonatal retina as a model for angiogenesis, I found that while BCL2 was not required for EC survival during angiogenic vessel growth (Chapter 4), MCL1 was required in a dose-dependent manner (Chapter 5). In contrast to normal angiogenesis, BCL2 and MCL1 were both independently required for the growth of abnormal vascular lesions in a murine model of pathological retina angiogenesis (Chapter 6). These studies have conclusively determined the role for EC apoptosis during angiogenic growth and remodelling and provide evidence that targeting distinct BCL2 family pro-survival proteins may be a useful therapeutic approach for targeting pathological angiogenesis.
  • Item
    Thumbnail Image
    The role of HECTD1 and MCL-1 in the regulation of normal and malignant haematopoiesis
    Brennan, Margs ( 2019)
    Cellular processes important for haematopoiesis are frequently perturbed in malignant cells. Accordingly, healthy immature progenitor cells and malignant cancer cells often share certain properties, e.g. rapid proliferation and self-renewal. Deciphering these developmental pathways can provide information about the critical drivers involved in neoplastic transformation and sustained cancer cell growth. Results in this thesis addresses the role of two proteins, HECTD1 and MCL-1, in haematopoiesis and haematological malignancies. HECTD1 is an E3 ubiquitin ligase required for mouse embryonic development, as homozygous loss of Hectd1 leads to embryonic lethality. HECTD1 is widely expressed in diverse tissues, including haematopoietic cells. However, its role in adult tissues in vivo has not been described. Therefore, we generated mice in which HECTD1 deletion was restricted to the haematopoietic system of adult mice. Analysis of these mice at steady state revealed small perturbations in certain T cell subsets. However, competitive reconstitution experiments revealed that HECTD1 deletion affects the haematopoietic stem and progenitor cell (HSPC) populations. Serial transplantation assays showed that loss of HECTD1 results in a defect in the self-renewal properties of mouse HSPCs. Interestingly, RNA sequencing of Hectd1-/- HSPCs revealed that HECTD1-deficiency led to increased expression of interferon regulated genes, suggesting that HECTD1 plays a critical role in the maintenance of HSPC populations by negatively regulating the interferon signalling pathway. Additionally, I employed the MLL AF9 mouse model of acute myeloid leukaemia and showed that HECTD1-deficiency significantly delayed the latency of tumour development in vivo compared to control mice. MCL-1 is a pro-survival regulator of the intrinsic apoptosis pathway. MCL-1 expression is integral to the survival of many different blood cell types, and to the development and sustained growth of many haematological malignancies. Recently a highly specific MCL-1 inhibitor, S63845, showing 6-fold higher affinity to human MCL-1 compared to mouse MCL-1 was described. To accurately test the efficacy and tolerability of S63845 in preclinical models of disease, we developed a humanised Mcl 1 (huMcl-1) mouse strain in which the genomic region of the murine Mcl-1 locus was replaced with the coding regions for human MCL-1. These mice are phenotypically indistinguishable from wild-type mice, and the intrinsic apoptotic pathway remains intact in their cells. However, as anticipated, huMcl-1 mice were more sensitive to S63845 than wild-type mice. To test whether malignant cells from the humanised MCL-1 mice also show higher sensitivity to S63845, we generated Eµ-Myc lymphomas on a huMcl-1 background. Lymphoma cell lines derived from huMcl-1;Eµ-Myc mice were ~6 times more sensitive to S63845 in vitro compared to Eµ-Myc lymphoma cells expressing mouse MCL-1. Transplantation of huMcl-1;Eµ-Myc lymphoma cells into huMcl-1 mice and treatment with S63845 resulted in tumour-free survival in >60% of mice. Furthermore, combining low doses of S63845 with sub-optimal doses of cyclophosphamide led to almost complete tumour regression. These results show that our huMcl-1 mouse model represents a valuable preclinical tool to test MCL-1 inhibitors, either alone or in combination with other anti-cancer agents, for a broad range of cancers, allowing accurate prediction of efficacy against tumour cells and on target toxicity to normal tissues.