The PhD in Biological Science (Stem Cell Biology) will be carried out under the supervision of a Principal Investigator (PI) from within the Cambridge Stem Cell Institute, and based in their research group.
Projects available for 2026-27 entry are listed below:
Join the Alcolea Group for a project on 'Mutant clone competition in squamous epithelial tissues; relevance for early tumour formation' (dry lab project).
Keywords: Mutant clonal competition, Early tumorigenesis, Squamous epithelia, Epithelial stem cells, Cell fate
Project Overview
Mounting evidence show that healthy human epithelial tissues accumulate cancer-associated mutations with age. However, we have very limited understanding as to how these mutations affect long-term tissue maintenance and contribute to early tumour formation.
This project will make use of an interdisciplinary approach combining computational approaches with clonal fate modelling to address the outstanding question of how changes in the mutational landscape affect cell fate dynamics in squamous epithelial tissues.
Work in this area will establish a baseline to understand how specific mutations impact cancer susceptibility as we grow older. The ultimate aim will be to identify potential strategies to modulate mutant cell behaviour and prevent cancer.
Main Techniques
In this dry lab project, we will combine our interdisciplinary approach to model epithelial fate dynamics in mutant cells. This will be achieved through single-cell multiomics, lineage tracing analysis and quantitative modelling. These will include:
- Analysis of sequencing-based data, particularly single-cell RNA sequencing. Other data may include e.g. WGS, ChIP-seq, ATAC-seq, bulk RNA-seq, multi-omics, and proteomics.
- Downstream data modelling, including pathway analysis, gene regulatory network analysis and cell fate modelling.
- Analysis of integrated multiomics data to decipher regulatory networks associated with mutant cells. Machine learning approached will enable enumerating and building of possible network topologies between cell types based on cell-cell interaction pairs to study cell communication.
- Quantitative lineage tracing analysis using statistical modelling-based approaches. As well as quantitative image analysis and large-scale image segmentation.
Composite created with images produced by members of the Alcolea Lab and collaborators.
Join the Alcolea Group for a project on 'Mutant clone competition in squamous epithelial tissues; relevance for early tumour formation' (wet lab project).
Keywords: Mutant clonal competition, Early tumorigenesis, Squamous epithelia, Epithelial stem cells, Cell fate
Project Overview
Mounting evidence show that healthy human epithelial tissues accumulate cancer-associated mutations with age, revealing that mutations do not represent the sole cause of cancer. Instead, tumour formation is now believed to depend on a combination of genetic and environmental factors; with competition between adjacent mutant clones acting as critical modulators during early tumorigenesis. However, despite significant efforts in the cancer field, we still know virtually nothing as to how mutant competition is affected by ageing, and whether this contributes to differences in tumour incidence with age.
This PhD project will address the question of how ageing impacts mutant clone dynamics to promote early tumour formation. In particular, we will focus on understanding how mutant clonal competition changes with age, from birth through to late stages in life, using a squamous oesophageal model.
Importantly, work in this area will provide a benchmark to identify new actionable targets of potential use in cancer therapeutics and diagnostics, and to develop strategies to prevent the onset of this aggressive disease.
Main Techniques
To dissect the cellular and molecular mechanisms underlying mutant competition and early tumorigenesis, we will implement a multidisciplinary approach combining our expertise in mutant clonal dynamics in vivo and ex vivo using 3D organ cultures, single-cell multiomics, and mathematical network analysis. Specific techniques will include:
- New long-term 3D Epitheliod cultures (Herms et al. BiorRxiv 2023).
- 3D organoid cultures.
- Clonal lineage tracing.
- Immunostaining of tissue wholemounts.
- 3D whole-organ confocal microscopy.
- Single-cell RNA sequencing and multiomics data mining.
- Gene Regulatory Networks will be assessed using the relevant models, including genetic manipulation via CRISPR/Cas9.
Composite created with images produced by members of the Alcolea Lab and collaborators.
Join the Bargehr Group for a project on 'Immunomodulatory strategies to optimise myocardial grafting efficiency following myocardial infarction'.
Keywords: Regenerative medicine, cardiac repair, immunomodulation, heart failure
Project Overview
Heart failure is the inability to sufficiently pump blood throughout the body’s circulation and 50% of patients diagnosed with it do not survive the first 5 years following their diagnosis. The heart’s contractile weakness is largely caused by ischaemic heart disease with an average heart attack resulting in the loss of approximately 1 billion cardiomyocytes. Cardiac repair has been using hPSC-derived cardiomyocytes to remuscularise infarcted hearts with robust grafts in rodents and large animals. However, a key limitation of this strategy is its inefficiency with most cells not resulting in long-term engraftment.
The Bargehr lab is exploring how the space between cardiac grafts and the host heart, which is largely dictated by the immune system, can be modulated to enhance cardiac grafting efficiency. This will include strategies to inhibit cytotoxic and NK cell responses and enhance the recruitment of regulatory T-cells to ameliorate inflammation and modify extracellular matrix deposition. This work will generate key insights into how cardiac grafting efficiency can be enhanced which will pave the way for translation of this technology to clinical trials.
Main Techniques
- Immunohistochemistry and confocal microscopy
- Single cell RNAseq analysis
- Spatial transcriptomics
- Generation of cardiomyocytes and epicardial cells from hPSC
- Myocardial cell transplantation & ex vivo heart perfusion
Infected rats transplanted with hPSC-derived cardiomyocytes. Human grafts depicted in green, electrical gap junctions in red. Image credit: Johannes Bargehr.
Join the Boroviak Group for a project on 'Engineering primate embryo implantation platforms'.
Keywords: human embryo implantation, primate embryogenesis, implantation platforms, endometrium
Project Overview
Embryo implantation is one of the most critical and complex events in early pregnancy. Central to this process is the establishment of a functional vascular network, which delivers oxygen and nutrients to support embryonic growth.
In this project, we will develop a vascularised endometrial implantation platform that mimics the spatial and functional organisation of the maternal–fetal interface to study primate embryogenesis ex vivo.
Embryo implantation requires the endometrium to undergo cyclical transformation. In humans and great apes, the endometrium undergoes cyclical transformations, including the decidualization of stromal cells and secretory changes of the glandular epithelium in response to progesterone. A key and often overlooked aspect of embryo implantation is vascularisation. Endometrial blood vessels, particularly spiral arteries, undergo extensive remodelling during early pregnancy, ensuring nutrient and oxygen delivery to the developing embryo. Yet current in vitro models insufficiently capture the spatial and functional complexity of this vascular network.
Our goal is to build a vascularised implantation platform. This bioengineered system integrates human stromal, endothelial and epithelial cells into a patterned hydrogel scaffold. The platform will feature two distinct compartments: (i) a vascularised stromal layer, in which microvessels spontaneously arise from embedded endothelial cells; and (ii) macrovascular channels created by 3D printing and perfused with endothelial cells to generate lumenised vessels. Upon assembly, the platform will be transferred into a microfluidic culture device that allows long-term perfusion. This setup supports the integration of micro- and macrovascular components and mimics physiological perfusion of the endometrium. Endothelial integration, hormonal responsiveness and tissue viability will be assessed using confocal imaging and spatial transcriptome profiling.
The student will learn endometrial organoid culture and differentiation, vascularised tissue bioprinting and microfluidic assembly, immunostaining, 3D imaging and image analysis, qPCR and gene expression analysis, as well as comparative primate developmental biology.
Ultimately, this project aims to recapitulate the critical features of the maternal-fetal interface and establish a foundation for modelling primate embryogenesis outside the womb.
References:
- Bergmann S, Schindler M, Munger C, Penfold CA, Boroviak TE. Building a stem cell-based primate uterus. Commun Biol. 2021.
- Siriwardena D, Boroviak TE. Evolutionary divergence of embryo implantation in primates. Phil Trans R Soc B. 2022.
- Rawlings TM et al. Modelling embryo implantation in human endometrial assembloids. Elife. 2021.
- Turco MY et al. Long-term, hormone-responsive organoid cultures of human endometrium. Nat Cell Biol. 2017.
- Zhang J et al. Modeling embryo-endometrial interface recapitulating human embryo implantation. Sci Adv. 2024. doi:10.1126/sciadv.adi4819
Main Techniques
- Organoid culture
- Generation of implantation platforms
- Confocal microscopy
- Single-cell transcriptome profiling
- Spatial transcriptome profiling.
Microvasculature embedded in human stromal and endometrial cells, generated in the Boroviak lab.
Join the Duque-Correa Group for a project on 'Interactions between whipworms and the stem cell niche that determine chronic infections'.
Keywords: Stem cell fate, intestinal stem cell niche, whipworm, parasitic nematode, infection
Project Overview
Whipworms (Trichuris trihiura) infect hundreds of millions of people and cause trichuriasis, a major neglected disease. These large metazoan parasites inhabit a multi-intracellular niche within their host gut lining, where they can remain for years.
Whipworm infection occurs upon ingestion of eggs that hatch in the caecum in a process mediated by the host microbiota. Motile first-stage L1 larvae released from the eggs transverse the mucus layers and enter the intestinal epithelia (IE) at the base of the crypts of Lieberkühn. Our research has shown that whipworm L1 larvae invade intestinal stem, deep secretory and progenitor cells (Duque-Correa et al 2022 Nature Communications); however, the impact of the parasite on the stem cell fate and stem cell niche remodelling remains unknown. To persist in their host for years, whipworms manipulate the IE renewal cycle (3-5 days) thus avoiding expulsion. Hence, we hypothesise that whipworms induce changes in stem cell division and differentiation as well as on the stromal populations of the stem cell niche, influencing IE architecture and composition in a manner that is critical to support parasite persistence in the host.
The aim of this PhD project is to identify critical interactions between whipworms and the intestinal epithelia, stromal and immune populations that result in remodelling of the stem cell niche to enable chronic infections.
We anticipate this work will lead to fundamental new knowledge on the whipworm mucosal niche, potentially yielding new targets for anti-parasitic therapies and providing novel insights into how intestinal epithelia repair damage with relevant application toward intestinal inflammatory diseases.
Main Techniques
To address this aim, you will use a novel organoid model developed by the Duque-Correa lab, the first to reproduce whipworm infection of the IE in vitro (Duque-Correa et al 2022 Nature Communications; Hofer et al 2025 Nature Biomedical Engineering), together with a mouse model of infection with the natural mouse whipworm (Trichuris muris). Using these models, you will:
- Identify changes in stem cell fate, stromal populations and tissue repair programmes induced by whipworm infection and key parasite molecules inducing these changes, using bulk, single-cell and spatial transcriptomics.
- Characterise stem cell niche remodelling and repair pathways modulated by the parasite using lineage tracing and confocal imaging of infected mice and assess stem and stromal cell function using organoid assays.
- Investigate the mechanisms by which candidate parasite molecules modulate stem cell niche remodelling.
Image Credit: https://doi.org/10.1038/s41467-022-29334-0
Join the Göttgens Group for a project on 'Decoding Stem Cell Fitness: Interpretable Gene Programs in Haematopoiesis and Ageing'.
Supervisors: Bertie Göttgens & Nicola Wilson
Project Overview
Single-cell transcriptomics has revolutionised our understanding of cellular diversity, but translating this complexity into mechanistic insights remains challenging. Traditional methods often reduce rich cellular states to single vectors, limiting interpretability and masking key regulatory processes.
This project builds on Tripso, a deep learning framework developed by the Lotfollahi group in collaboration with the Gottgens lab. Tripso uses gene program (GP)-specific transformers to learn multiple interpretable representations of gene activity at the single-cell level. Applied to over 500,000 haematopoietic cells, Tripso revealed age-specific GP usage and uncovered novel programs driving cell fate decisions and stem cell fitness.
A core aim is experimental validation of GP-based hypotheses. The student will collaborate with computational researchers to identify candidate programs and targets, testing predictions via CRISPR perturbations, pharmacological treatments, and flow cytometry. Functional outcomes will be assessed using molecular characterisation techniques, building on prior success such as Tripso’s prediction that SEC61 inhibition enhances stem cell maintenance.
This interdisciplinary project bridges computational modelling and experimental haematology. The student will gain training in single-cell transcriptomics, machine learning (including transformers and optimal transport), and hands-on lab techniques. They will learn to generate hypotheses from large-scale data and design experiments to test gene program function across developmental stages and ageing contexts.
Ultimately, the project aims to uncover mechanisms regulating stem cell fitness, ageing, and lineage commitment, with implications for regenerative medicine and stem cell therapies.
Join the Duque-Correa Group for a project on 'Intestinal stem cell niche: a home for whipworms to develop'.
Keywords: Intestinal stem cell niche, whipworm, parasitic nematode, infection, worm development
Project Overview
Human whipworms (Trichuris trihiura) infect hundreds of millions of people and cause trichuriasis, a major neglected disease. These large metazoan parasites inhabit a multi-intracellular niche within the human gut lining, where they can remain for years. Specific tissue adaptations unique to whipworms enable their intracellular life-style and are likely the result of co-evolutionary processes between these parasites and their host intestinal stem cell niche. The development of those tissues across the parasite life-cycle and the host signals/factors triggering whipworm developmental trajectories are not understood. The aim of this PhD project is to determine the molecular and morphological changes underlying whipworm development and the function of critical parasite tissues that support its survival within the host intestinal epithelia.
Main Techniques
To address this aim, you will exploit a novel organoid model developed by the Duque-Correa lab, the first to reproduce whipworm infection of the intestinal epithelia in vitro (Duque-Correa et al 2022 Nature Communications; Hofer et al 2025 Nature Biomedical Engineering), together with a mouse model of infection with the natural mouse whipworm (Trichuris muris). Using these models, you will:
- Characterise the developmental trajectories and metabolic requirements of whipworms through their life cycle via bulk, single-cell and spatial RNA-sequencing.
- Identify key host factors and regulators of parasite development, using advanced imaging and transcriptomics.
- Determine the ontogeny and function of whipworm tissues that enable parasite intracellular living.
Join the Káradóttir Group for a project on 'Determining mechanism of progression in MS'.
Keywords: CNS, myelin, Oligodendrocyte, neuroscience, stem cells, brain, Multiple Sclerosis
Project Overview
Myelin is essential for normal brain function. Throughout life ligodendrocyte precursor cells (OPCs) which are stem cells in the brain and the main proliferative cells in the adult central nervous system, differentiate into myelinating oligodendrocytes to maintain normal brain function. While OPCs in young animals effectively differentiate and respond to demyelinating lesions as occurs in multiple sclerosis (MS). However, with ageing their differentiation capacity declines, which thought to be the main contributor to progressive disability in diseases like MS. Recent genome-wide association studies have identified a genetic variant that accelerates MS progression, with carriers reaching severe disability years earlier and showing greater brain atrophy and lesion burden.
We are establishing a humanised mouse model for this gene variant to investigate how it affects OPC biology and myelin regeneration. Preliminary single-cell analyses suggest this variant causes vulnerability in oligodendrocytes with emerging senescence signatures in glial cells. This studentship will characterise the stem cell regenerative programme in these mice, examining OPC proliferation, differentiation, and remyelination capacity to identify the cellular and molecular mechanisms underlying accelerated disease progression in genetic variant carriers.
Main Techniques
- confocal imaging
- in vivo photometry
- stereotaxic surgery
- histology
Image shows: Microglia (red) hugging demyelinating neuron (in blue). Image credit: Omar de Faria
Join the Káradóttir Group for a project on 'Identifying the cell state that drives myelin repair'.
Keywords: CNS, myelin, oligodendrocyte, neuroscience, stem cells, brain, multiple sclerosis, remyelination
Project Overview
Oligodendrocyte progenitor cells (OPCs) constitute a resident stem cell population in the central nervous system, maintaining both their proliferation capacity as well as their ability to differentiate into oligodendrocytes, the cells that make myelin, a substance necessary for normal brain function. Myelin can be damaged in a variety of conditions, such as multiple sclerosis (MS). Following demyelination, OPCs are the cells responsible for responding and repairing the lost myelin. It is known however, that OPCs are not a homogenous cell population, but they exist in different cell states.
We have established an interdisciplinary approach to identify and characterise different OPC states both during normal brain development/ageing, as well as following focal myelin damage. This project aims to identify the OPC state that is more competent for remyelination, by performing clonal analysis in demyelinating lesions and combining histology with transcriptomic analysis to molecularly describe this regenerative OPC state.
Main Techniques
- Stereotactic surgery
- lineage tracing
- histology
- confocal imaging
- transcriptomic analysis
Image shows: Oligodendrocyte progenitor cells, coloured by different z-plane. Image credit: Stavros Vagionitis
Join the Khaled Group for a project on 'Understanding and intercepting precancerous cellular changes in the breast'.
Keywords: Tumour initiation, Cancer Prevention, lineage tracing
Project Overview
Understanding the molecular and cellular mechanisms of how epithelial tissues maintain a homeostatic state throughout the lifespan of mammals is a major challenge for developmental and stem cell biology. From a developmental perspective, the epithelium of the mammary gland is unique as it undergoes most of its development during adulthood. Despite recent efforts of characterising the tissue homeostasis at a cellular level little is known about how this is affected by various developmental processes such as pregnancy or ageing and how this might ultimately disrupt epithelial homeostasis resulting in malignant outgrowth. In this study we wish to further our understanding of the changing nature of the differentiation dynamics of the mammary gland. To fully understand how tissue homeostasis is affected by parity and other events it is mandatory to characterise the differentiation dynamics in an age-dependent manner. This becomes evident when looking at epidemiological data. Age is the greatest risk factor for breast carcinomas, and it has been suggested that this is not only due to accumulation of mutations but also due to decreased clonality and selection of clones with proliferative advantage. To tackle these questions our lab has been using single cell genomics, mouse models and human samples to study the effects of parity, ageing and germline mutation (BRCA1/2) on the homeostasis of the mammary gland in mouse and human. For this project the student will be using a new lentiviral system we developed in the lab that is designed to induce and facilitate comprehensive characterisation of dynamic changes in the mammary epithelium of Cre-inducible mouse models, such as confetti and tumour suppressor flox mouse models. In addition, the student will have the opportunity to validate some of the findings using orthogonal spatial technologies as well as perform pre-clinical in vivo cancer interception studies.
Main Techniques
- Spatial transcriptomics
- Single cell genomics
- Tissue scale imaging
- Mouse models
Join the Teichmann Group for a project on 'Cardiac Cell Circuits – deciphering human heart function'.
Keywords: Spatial transcriptomics of the heart
Project Overview
To define the molecular map of human cardiac tissues, we are applying cell atlas technologies to decipher the molecular mechanism of cell-cell communication in the dozens of tissue niches comprising the human heart. This will contribute to assembling the complete human heart cell atlas, leading to deeper understanding of the function of this organ and its paracrine, endocrine and neural signalling circuits. We apply technologies such as Xenium, VisiumHD and large-volume imaging in an integrative approach to probe novel aspects of heart biology.
We use modelling and machine learning to define a Common Coordinate Framework (CCF) in an Organ-Axis approach (Yayon, Kedlian, Boehme et al, Nature, in press), which could be applied to the lumen-adventitia axis of the big arteries in the heart. Features like cell type abundance or nearest neighbours can then be compared for all three technologies along this axis. Similarly, we will compare tissue niches defined by different technologies and examine if higher resolution technologies can ‘decompose’ the niches with a view to gaining new insights into the cell type composition and cell signalling circuits relevant in heart function.
Main Techiques
Computational analysis of existing data, plus new data generation using cutting edge spatial transcriptomics technology (eg VisiumHD, StereoSeq, Slide-tag)
Image credit: Kazumasa Kanemaru, James Cranley
Join the Teichmann Group for a project on 'Multiomics to decipher T cell development in the thymus'.
Keywords: T cell development
Project Overview
Ongoing projects in the Teichmann team are generating scRNA-seq and chromatin accessibility profiles of thymic cells using the 10X multiome kit, combined with multiplex protein staining. The project will examine how altered chromatin states might direct specific T cell fates, examining both T cells and thymic epithelial cells in fetal and paediatric thymi. We have established artificial thymic organoids (ATO) culture systems and will test our insights from single cell data to modulate T cell development in vitro via overexpression of specific transcription factors (TFs) to direct T cell fates. Computational models will be harnessed to predict which TFs may be essential to drive different effector T cell fates.
Main Techniques
- Computational analysis of existing data, plus new data generation using cutting edge TCR repertoire analysis.
- Thymic organoid culture and transfections.
Image credit: Ioannis Sarropoulos
Join the Teichmann Group for a project on 'Deciphering the human T cell receptor code'.
Keywords: T cell repertoires
Project Overview
Our laboratory is interested in understanding the establishment of T cell receptor repertoires and the role played by polymorphic peptide-MHC complexes in shaping this. This project will examine recurring motifs in paired alpha and beta chain TCR sequences, and model these with tools such as AlphaFold and utilise deep learning approaches to try to predict binding specificities. Extensive TCR sequencing from single cell data will also from the basis for developing better tools to understand the VDJ rearrangement process, including the identification of D-D, reversed or dual recombination on top of non-functional rearrangements to understand whether these are part of productive TCRs and how these TCRs are distributed within the range of T cell subtypes. This will form the basis for linking TCR motifs to their cognate peptide-MHC targets using data mining of unpublished and public datasets.
Main Techniques
- Computational analysis of existing data, plus new data generation using cutting edge approaches for single cell genomics paired chain TCR repertoire analysis.
- Deep learning and AI tools for structure predictions.
Image credit: Yizhou Yi
Join the Teichmann Group for a project on 'Deciphering gut microbe-immune interactions in health and disease'.
Keywords: Gut, immunology, microbiome, T cells
Project Overview
A fundamental role of our immune system is to defend against harmful threats, whilst minimising immune-mediated pathologies. The maintenance of immune tolerance across barrier surfaces such as the gut where the self interfaces with the microbial non-self represents unique challenges to the mucosal immune system. The gut is colonised by a large community of commensal bacteria that contributes to health through a complex dialogue with the host to promote tolerance and defence.
The study of microbe-immune crosstalk is therefore highly relevant to understand how tolerance is broken in pathologies such as inflammatory bowel disease and gut graft versus host disease. This project incorporates 3 broad aims:
1. Develop spatial tools to analyse high-resolution spatial transcriptomic gut tissue data
2. Generate spatial data to investigate host-microbe interactions using human gut biopsies across the spectrum of health and disease
3. Use iPSC and patient-derived intestinal organoid lines established in our group to test new mechanisms by which the host co-exists with microbes.
Main Techniques
- Computational analysis of existing data
- Generation of new data generation using cutting edge spatial transcriptomics technology (e.g., VisiumHD)
- Intestinal organoid culture
Generation of human intestinal organoids from induced pluripotent stem cells (iPSC). Image credit:
Join the Tyser Group for a project on 'Examining the interplay between form and function during heart development'.
Keywords: Heart Development, Physiology
Project Overview
The heart is the first organ to form and function during development, essential in providing the embryo with sufficient oxygen and nutrients. During heart development, multipotent cardiac progenitors undergo a process of differentiation, whilst also initiating and maintaining contraction. The forming heart is therefore a good model to explore the relationship between function and differentiation/form. Our group is interested in how cardiac function may act as a signalling component to regulate differentiation and heart formation. This work will provide insight into the origins of congenital heart disease and contribute mechanistic understanding as to how the reactivation of developmental gene programs impacts heart disease.
Main Techniques
- Whole mount and Timelapse imaging
- Mammalian embryology and dissection
- Single cell RNAseq analysis.
- hESC cardiac differentiation.
Wholemount immunofluorescent image of the developing mouse heart at embryonic day 12.5. Muscle is shown in red and nuclei (DAPI) in grey. Image credit: Richard Tyser.
Join the Tzelepis Group for a project on 'Identification of pathways regulating the presentation of csRBP-glycoRNA clusters on the surface of mammalian cells'.
Keywords: RNA biology, Epitranscriptomics, stem cells
Project Overview
Immunotherapies for acute myeloid leukemia (AML) and other cancers are limited by a lack of tumor-specific targets. We recently discovered that RNA-binding proteins and glycosylated RNAs (glycoRNAs) form precisely organized nanodomains on cancer cell surfaces. We characterized csNPM1 (George et al, Nature Biotechnology 2025) and csDDX21 (Perr et al, Cell 2025) as abundant cell surface proteins on a variety of tumor types. We develop a monoclonal antibody to target csNPM1 and csDDX21, which exhibit robust anti-tumor activity in multiple syngeneic and xenograft models of AML, including patient-derived xenografts, without observable toxicity. Our data suggest that csNPM1 and its neighboring glycoRNA–cell surface RNA-binding protein (csRBP) clusters may serve as an alternative antigen class for therapeutic targeting or cell identification.
For this project, we have performed genome-wide CRISPR screens and have catalogued gene targets that changed the levels of the above glycoRNA–cell surface RNA-binding protein (csRBP) clusters. Using sophisticated functional genetic and molecular biology techniques, we are aiming to validate and characterise the most promising targets we have recently identified, that either increased or decreased the presence of glycoRNA–cell surface RNA-binding protein (csRBP) clusters on the surface of normal and malignant models.
Main Techniques
- Cell culture
- CRISPR editing
- flow cytometry
- western blotting
- cell imaging
- There is also potential to perform translational studies using mouse models of disease that are established in the lab.
Join the Vassiliou Group for a project on 'Telomere maintenance, ageing and leukaemia prevention'.
Keywords: Clonal haematopoiesis, telomeres, leukaemia prevention
Project Overview
Acute myeloid leukaemia (AML) and related myeloid cancers develop from the premalignant phenomenon of clonal haematopoiesis (CH), the clonal expansion of a haematopoietic stem cell (HSC) and its progeny driven by somatic driver mutations. The vast majority of cases of CH are driven by mutations affecting a specific set of genes, namely epigenetic regulators DNMT3A, TET2 & ASXL1, splicing factor genes SRSF2, SF3B1 & U2AF1, DNA damage response genes TP53 & PPM1D and the signalling kinase gene JAK2. Individuals at high risk of progression from CH to myeloid cancer can be identified years in advance from the nature of the somatic mutations and the clonal size of CH. Our team is working to improve our understanding of how CH develops and how it can be prevented from progressing to myeloid cancer.
Recently, we and others recently reported that people who inherit genetic variants associated with longer telomeres are at increased risk of developing CH. Our downstream investigations of these findings are revealing that telomere length and maintenance are critical modulators of CH development and progression. In fact we are finding that whilst some types of CH thrive in people with long telomeres, others develop in individuals with shorter telomeres. Our findings reveal that telomere maintenance is critical to CH expansion and that established CH may be vulnerable to interference with telomere maintenance. If this is confirmed it has the potential to help us develop therapeutic interventions to reverse or curtail CH expansion and help prevent the development of myeloid cancers in those at risk.
The interaction between telomere maintenance and CH is intricately linked to ageing. During cell division, the DNA replication machinery cannot copy the very end of each chromosomal telomere, a concept known as “the end replication problem”. This results in loss of 50-100 base pairs of telomeric DNA with each cell division. As a result, telomeres shorten with successive cell divisions and eventually reach a critical threshold that triggers the DNA damage response and induces replicative senescence, preventing further cell divisions. Normal HSCs express telomerase (TERT), a specialised reverse transcriptase that adds telomere repeat sequences to the end of telomeres allowing HSCs to divide and replicate their DNA multiple times. Despite expressing TERT, even HSCs lose telomeric DNA with time, leading to impaired proliferative and tissue regeneration potential. It is in this context that somatic mutations engender CH and as a result different types of CH expand at different ages.
The successful applicant will investigate the basis of the interaction between one of the different types of CH, ageing and telomere maintenance. Our lab is interested in all types of CH and the precise focus of the project will be developed to suit the interests and skills of the successful candidate. There will be opportunity to use experimental studies in humans and mice, computational approaches, next generation sequencing, analysis of data from large cohorts such as the UK Biobank and other omics approaches.
Main Techniques
There will be opportunity to use experimental studies in humans and mice, computational approaches, next generation sequencing (including long-read and single cell), analysis of data from large cohorts such as the UK Biobank and other omics approaches.
Join the Zilbauer Group for a project on 'Epigenetic Regulation and Disease Modelling Using Human Intestinal Organoids'.
Keywords: Intestinal organoids, epigenetics, DNA methylation, IBD, stem cell biology, translational medicine, gene editing, computational biology, AI, Machine Learning
Project Overview
This PhD project will investigate the molecular mechanisms that govern intestinal epithelial cell function in health and disease, with a particular focus on epigenetic regulation. The successful candidate will work with human, mucosa-derived intestinal organoids to explore how DNA methylation and other epigenetic mechanisms contribute to epithelial cell identity, memory, and dysfunction in Inflammatory Bowel Disease (IBD).
A key area of development will be the establishment and application of advanced co-culture systems, integrating epithelial, mesenchymal and immune cells as well as the microbiome to model complex human intestinal tissue environments. Furthermore, we will apply cutting edge gene editing techniques to a range of human gut organoids and subject them to molecular as well as functional assays.
The Zilbauer lab leads an internationally recognised translational research programme applying multi-omic approaches to large patient-derived organoid biobanks. We collaborate with multiple clinical and academic centres across the UK and Europe, and the project will benefit from access to one of the largest human gut organoid biobanks in the world, containing over 1,500 deeply profiled and characterised lines.
Current areas of interest include the development of a molecular classification system for IBD, epigenetic biomarker discovery, and epithelial-targeted therapeutic strategies.
The exact project will be tailored to the student’s background and interests, and may include a combination of wet-lab work and computational analysis. Applicants from both biomedical and quantitative disciplines are encouraged to apply. We are a highly interdisciplinary group with close links to the clinical team at Cambridge University Hospitals.
Main Techniques
- Human intestinal organoid culture
- Development of co-culture systems (epithelium + mesenchyme + immune cells)
- Genome-wide DNA methylation and RNA sequencing (bulk/scRNAseq/ATACseq etc)
- Gene editing (e.g. CRISPR-Cas9)
- Cytokine stimulation assays
- High throughput imaging
- Computational biology and multi-omics integration (AI, ML)
Human intestinal epithelial organoids. Image credit: Tom Dennison