Forthcoming events in this series


Fri, 10 Mar 2023

14:00 - 15:00
Virtual

CRISPR-based decoding of disease-associated genomic variants

Prof Richard Sherwood
(Brigham and Womens Hospital Harvard Medical School)
Abstract

The overall goal of the Sherwood lab is to advance genomic and precision medicine applications through high-throughput, multi-disciplinary science. In a shortened talk this past autumn, I described our recent efforts using combined analysis of rare coding variants from the UK Biobank and genome-scale CRISPR-Cas9 knockout and activation screening to improve the identification of genes, coding variants, and non-coding variants whose alteration impacts serum LDL cholesterol (LDL-C) levels.

In this talk, I will discuss our emerging efforts to optimize and employ precision CRISPR techniques such as base editing and prime editing to better understand the impacts of coding and non-coding variation on serum LDL-C levels and coronary artery disease risk. This work involves the development of novel high-throughput screening platforms and computational analysis approaches that have wide applicability in dissecting complex human disease genetics.

Fri, 03 Mar 2023

14:00 - 15:00
Virtual

An agent-based model of the tumour microenvironment

Dr Cicely Macnamara
(School of Mathematics and Statistics University of Glasgow)
Abstract

The term cancer covers a multitude of bodily diseases, broadly categorised by having cells which do not behave normally. Cancer cells can arise from any type of cell in the body; cancers can grow in or around any tissue or organ making the disease highly complex. My research is focused on understanding the specific mechanisms that occur in the tumour microenvironment via mathematical and computational modelling. In this talk I shall present a 3D individual-based force-based model for tumour growth and development in which we simulate the behaviour of, and spatio-temporal interactions between, cells, extracellular matrix fibres and blood vessels. Each agent is fully realised, for example, cells are described as viscoelastic sphere with radius and centre given within the off-lattice model. Interactions are primarily governed by mechanical forces between elements. However, as well as he mechanical interactions we also consider chemical interactions, by coupling the code to a finite element solver to model the diffusion of oxygen from blood vessels to cells, as well as intercellular aspects such as cell phenotypes. 

Fri, 24 Feb 2023

14:00 - 15:00
L3

Decoding nanopore signals

Dr David Page
(Oxford Nanopore Technologies plc)
Abstract

Nanopore sequencing is a method to infer the sequence of nucleotides in DNA or RNA molecules from small variations in ionic current during transit through a nanoscale pore. We will give an introduction to nanopore sequencing and some of its applications and then explore simple models of the signal generation process. These can provide insight to guide optimisation of the system and inform the design of more flexible neural network models, capable of extracting the rich contextual information required for accurate sequence inference.

Fri, 17 Feb 2023

14:00 - 15:00
L3

Understanding Turing patterns in heterogeneous reaction-diffusion systems

Prof Robert A. Van Gorder
(Department of Mathematics and Statistics University of Otago)
Abstract

The Turing pattern is a key concept in the modern study of reaction-diffusion systems, with Turing patterns proposed a possible explanation for the spatial structure observed in myriad physical, chemical, and biological systems. Real-world systems are not always so clean as idealized Turing systems, and in this talk we will take up the case of more messy reaction-diffusion systems involving explicit space or time dependence in diffusion or reaction terms. Turing systems of this nature arise in several applications, such as when a Turing system is studied on a growing substrate, is subjected to a temperature gradient, or is immersed within a fluid flow. The analysis of these messy Turing systems is not as straightforward as in the idealized case, with the explicit space or time dependence greatly complicating or even preventing most standard routes of analysis. Motivated by patterning in phenomena involving explicit space or time dependence, and by the interesting mathematical challenges inherent in the study of such systems, in this talk we consider the following questions:

* Is it possible to obtain generalizations of the Turing instability conditions for non-autonomous, spatially heterogeneous reaction-diffusion systems?
* What can one say about predicting nascent patterns in these systems?
* What role does explicit space or time dependence play in selecting fully developed patterns in these systems?
* Is it possible to exploit this space or time dependence in order to manipulate the form of emergent patterns?

We will also highlight some of the applications opened up by the analysis of heterogeneous Turing systems.

Fri, 10 Feb 2023

14:00 - 15:00
L3

Inference of stem cell and tissue dynamics in development and regeneration

Dr Linus Schumacher
(Centre For Regenerative Medicine University of Edinburgh)
Abstract

The dynamics of a tissue in development or regeneration arises from the behaviour of its constituent cells and their interactions. We use mathematical models and inference from experimental data to to infer the likely cellular behaviours underlying changing tissue states. In this talk I will show examples of how we apply canonical birth-death process models to novel experimental data, how we are extending such models with volume exclusion and multistate dynamics, and how we attempt to more generally learn cell-cell interaction models directly from data in interpretable ways. The applications range from in vitro models of embryo development to in vivo blood regeneration that is disrupted with ageing.

Fri, 03 Feb 2023

14:00 - 15:00
L3

Challenges in modeling the transmission dynamics of childhood diseases

Prof Felicia Magpantay
(Dept of Math and Stats Queen’s University Kingston)
Abstract

Mathematical models of childhood diseases are often fitted using deterministic methods under the assumption of homogeneous contact rates within populations. Such models can provide good agreement with data in the absence of significant changes in population demography or transmission, such as in the case of pre-vaccine era measles. However, accurate modeling and forecasting after the start of mass vaccination has proved more challenging. This is true even in the case of measles which has a well understood natural history and a very effective vaccine. We demonstrate how the dynamics of homogeneous and age-structured models can be similar in the absence of vaccination, but diverge after vaccine roll-out. We also present some fundamental differences in deterministic and stochastic methods to fit models to data, and propose techniques to fit long term time series with imperfect covariate information. The methods we develop can be applied to many types of complex systems beyond those in disease ecology.
 

Fri, 27 Jan 2023

14:00 - 15:00
Virtual

Digital twin models for the precision diagnosis and therapy of cancer

Prof Walter Kolch
(School of Medicine University College Dublin)
Abstract

Approaches to personalized diagnosis and treatment in oncology are heavily reliant on computer models that use molecular and clinical features to
characterize an individual patient’s disease. Most of these models use genome and/or gene expression sequences to develop classifiers of a patient’s
tumor. However, in order to fully model the behavior and therapy response of a tumor, dynamic models are desirable that can act like a Digital Twin of
the cancer patient allowing prognostic and predictive simulations of disease progression, therapy responses and development of resistance. We are
constructing Digital Twins of cancer patients in order to perform dynamic and predictive simulations that improve patient stratification and
facilitate the design of individualized therapeutic strategies. Using a hybrid approach that combines artificial intelligence / machine learning
with dynamic mechanistic modelling we are developing a computational framework for generating Digital Twins. This framework can integrate
different types of data (multiomics, clinical, and existing knowledge) and produces personalized computational models of a patient’s tumor. The
computational models are validated and refined by experimental work and in retrospective patient studies. We present some of the results of the dynamic
Digital Twins simulations in neuroblastoma. They include (i) identification on non-MYCN amplified high risk patients; (ii) prediction of individual
patients’ responses to chemotherapy; and (iii) identification of new drug targets for personalized therapy. Digital Twin models allow the dynamic and
mechanistic simulation of disease progression and therapy response. They are useful for the stratification of patients and the design of personalized
therapies.

Fri, 20 Jan 2023

14:00 - 15:00
L3

The inevitable emergence of density-dependent diffusion in expanding phage populations

Dr Diana Fusco
(Dept of Physics University of Cambridge)
Abstract

Reaction-diffusion waves have long been used to describe the growth and spread of populations undergoing a spatial range expansion. Such waves are generally classed as either pulled, where the dynamics are driven by the very tip of the front and stochastic fluctuations are high, or pushed, where cooperation in growth or dispersal results in a bulk-driven wave in which fluctuations are suppressed. These concepts have been well studied experimentally in populations where the cooperation leads to a density-dependent growth rate. By contrast, relatively little is known about experimental populations that exhibit a density-dependent dispersal rate.

Using bacteriophage T7 as a test organism, we present novel experimental measurements that demonstrate that the diffusion of phage T7, in a lawn of host E. coli, is hindered by steric interactions with host bacteria cells. The coupling between host density, phage dispersal and cell lysis caused by viral infection results in an effective density-dependent diffusion rate akin to cooperative behavior. Using a system of reaction-diffusion equations, we show that this effect can result in a transition from a pulled to pushed expansion. Moreover, we find that a second, independent density-dependent effect on phage dispersal spontaneously emerges as a result of the viral incubation period, during which phage is trapped inside the host unable to disperse. Our results indicate both that bacteriophage can be used as a controllable laboratory population to investigate the impact of density-dependent dispersal on evolution, and that the genetic diversity and adaptability of expanding viral populations could be much greater than is currently assumed.

Fri, 18 Nov 2022

14:00 - 15:00
L3

Beyond DNA damage

Prof Hooshang Nikjoo
(Department of Physiology Anatomy & Genetics, University of Oxford )
Fri, 11 Nov 2022

14:00 - 15:00
L3

Identifying cell-to-cell variability using mathematical and statistical modelling

Dr Alex Browning
(Dept of Mathematics, University of Oxford)
Abstract

Cell-to-cell variability is often a primary source of variability in experimental data. Yet, it is common for mathematical analysis of biological systems to neglect biological variability by assuming that model parameters remain fixed between measurements. In this two-part talk, I present new mathematical and statistical tools to identify cell-to-cell variability from experimental data, based on mathematical models with random parameters. First, I identify variability in the internalisation of material by cells using approximate Bayesian computation and noisy flow cytometry measurements from several million cells. Second, I develop a computationally efficient method for inference and identifiability analysis of random parameter models based on an approximate moment-matched solution constructed through a multivariate Taylor expansion. Overall, I show how analysis of random parameter models can provide more precise parameter estimates and more accurate predictions with minimal additional computational cost compared to traditional modelling approaches.

Fri, 28 Oct 2022

14:00 - 15:00
L3

Emergent digital biocomputation through spatial diffusion and engineered bacteria

Prof Chris Barnes
(Dept of Cell and Developmental Biology UCL) )
Abstract

Building computationally capable biological systems has long been an aim of synthetic biology. The potential utility of bio-computing devices ranges from biosafety and environmental applications to diagnosis and personalised medicine. Here we present work on the design of bacterial computers which use spatial patterning to process information. A computer is composed of a number of bacterial colonies which, inspired by patterning in embryo development, communicate using diffusible morphogen-like signals. A computation is programmed into the overall physical arrangement of the system by arranging colonies such that the resulting diffusion field encodes the desired function, and the output is represented in the spatial pattern displayed by the colonies. We first mathematically demonstrate the simple digital logic capability of single bacterial colonies and show how additional structure is required to build complex functions. Secondly, inspired by electronic design automation, an algorithm for designing optimal spatial circuits computing two-level digital logic functions is presented, extending the capability of our system to complex digital functions without significantly increasing the biological complexity. We implement experimentally a proof-of-principle system using engineered Escherichia coli interpreting diffusion fields formed from droplets of an inducer molecule. Our approach will open up new ways to perform biological computation, with applications in synthetic biology, bioengineering and biosensing. Ultimately, these computational bacterial communities will help us explore information processing in natural biological systems.

Fri, 21 Oct 2022

14:00 - 15:00
L3

Systematic elucidation of genetic mechanisms underlying cholesterol uptake

Prof ~Richard Sherwood
(Brigham and Womens Hospital Harvard Medical School)
Abstract

The overall goal of the Sherwood lab is to advance genomic and precision medicine applications through high-throughput, multi-disciplinary science. In this talk, I will review a suite of high-throughput genomic and cellular perturbation platforms using CRISPR-based genome editing that the lab has developed to improve our understanding of genetic disease, gene regulation, and genome editing outcomes.

This talk will focus on recent efforts using combined analysis of rare coding variants from the UK Biobank and genome-scale CRISPR-Cas9 knockout and activation screening to improve the identification of genes, coding variants, and non-coding variants whose alteration impacts serum LDL cholesterol (LDL-C) levels. Through these efforts, we show that dysfunction of the RAB10 vesicle transport pathway leads to hypercholesterolemia in humans and mice by impairing surface LDL receptor levels. Further, we demonstrate that loss of function of OTX2 leads to robust reduction in serum LDL-C levels in mice and humans by increasing cellular LDL-C uptake. Finally, we unveil an activity-normalized base editing screening framework to better understand the impacts of coding and non-coding variation on serum LDL-C levels, altogether providing a roadmap for further efforts to dissect complex human disease genetics.

Fri, 14 Oct 2022
14:00
L3

Stochastic dynamics of cell fate decisions and the gene regulatory networks that underlie them

Prof Adam MacLean
(USC Dornsife College of Letters, Arts and Sciences University of Southern California)
Abstract

Cell fate decision-making is responsible for development and homeostasis, and is dysregulated in disease. Despite great promise, we are yet to harness the high-resolution cell state information that is offered by single-cell genomics data to understand cell fate decision-making as it is controlled by gene regulatory networks. We describe how we leveraged joint dynamics + genomics measurements in single cells to develop a new framework for single-cell-informed Bayesian parameter inference of Ca2+ pathway dynamics in single cells. This work reveals a mapping from transcriptional state to dynamic cell fate. But no cell is an island: cell-internal gene regulatory dynamics act in concert with external signals to control cell fate. We developed a multiscale model to study the effects of cell-cell communication on gene regulatory network dynamics controlling cell fates in hematopoiesis. Specifically, we couple cell-internal ODE models with a cell signaling model defined by a Poisson process. We discovered a profound role for cell-cell communication in controlling the fates of single cells, and show how our results resolve a controversy in the literature regarding hematopoietic stem cell differentiation. Overall, we argue for the need to consider single-cell-resolved models to understand and predict the fates of cells.

Fri, 17 Jun 2022

14:00 - 15:00
L6

Data-driven early detection of potential high risk SARS-CoV-2 variants

Dr Marcin J. Skwark
(InstaDeep)
Abstract

Recent advances in Deep Learning have enabled us to explore new application domains in molecular biology and drug discovery - including those driven by complex processes that defy analytical modelling.  However, despite the combined forces of increased data, improving compute resources and continuous algorithmic innovation all bringing previously intractable problems into the realm of possibility, many advances are yet to make a tangible impact for life science discovery.  In this talk, Dr Marcin J. Skwark will discuss the challenge of bringing machine learning innovation to tangible real-world impact.  Following a general introduction of the topic, as well as newly available methods and data, he will focus on the modelling of COVID-19 variants and, in particular, the DeepChain Early Warning System (EWS) developed by InstaDeep in collaboration with BioNTech.  With thousands of new, possibly dangerous, SARS-CoV-2 variants emerging each month worldwide, it is beyond humanities combined capacity to experimentally determine the immune evasion and transmissibility characteristics of every one.  EWS builds on an experimentally tested AI-first computational biology platform to evaluate new variants in minutes, and is capable of risk monitoring variant lineages in near real-time.  This is done by combining an AI-driven protein structure prediction framework with large, spike protein sequence-oriented Transformer models to allow for rapid simulation-free assessment of the immune escape risk and expected fitness of new variants, conditioned on the current state of the world.  The system has been extensively validated in cooperation with BioNTech, both in terms of host cell infection propensity (including experimental assays of receptor binding affinity), and immune escape (pVNT assays with monoclonal antibodies and real-life donor sera). In these assessments, purely unsupervised, data-first methods of EWS have shown remarkable accuracy. EWS flags and ranks all but one of the SARS-CoV-2 Variants of Concern (Alpha, Beta, Gamma, Delta… Omicron), discriminates between subvariants (e.g. BA.1/BA.2/BA.4 etc. distinction) and for most of the adverse events allows for proactive response on the day of the observation. This allows for appropriate response on average six weeks before it is possible for domain experts using domain knowledge and epidemiological data. The performance of the system, according to internal benchmarks, improves with time, allowing for example for supporting the decisions on the emerging Omicron subvariants on the first days of their occurrence. EWS impact has been notable in general media [2, 3] for the system's applicability to a novel problem, ability to derive generalizable conclusions from unevenly distributed, sparse and noisy data, to deliver insights which otherwise necessitate long and costly experimental assays.

Fri, 10 Jun 2022

14:00 - 15:00
L6

Systems-mechanobiology of health and disease

Dr Fabian Spill
(School of Mathematics University of Birmingham)
Abstract

Experimental biologists study diseases mostly through their abnormal molecular or cellular features. For example, they investigate genetic abnormalities in cancer, hormonal imbalances in diabetes, or an aberrant immune system in vascular diseases. Moreover, many diseases also have a mechanical component which is critical to their deadliness. Most notably, cancer kills typically through metastasis, where the cancer cells acquire the capability to remodel their adhesions and to migrate. Solid tumours are also characterised by physical changes in the extracellular matrix – the material surrounding the cells. While such physical changes are long known, only relatively recent research revealed that cells can sense altered physical properties and transduce them into chemical information. An example is the YAP/TAZ signalling pathway that can activate in response to altered matrix mechanics and that can drive tumour phenotypes such as the rate of cell proliferation.
Systems-biology models aim to study diseases holistically. In this talk, I will argue that physical signatures are a critical part of many diseases and therefore, need to be incorporated into systems-biology. Crucially, physical disease signatures bi-directionally interact with molecular and cellular signatures, presenting a major challenge to developing such models. I will present several examples of recent and ongoing work aimed at uncovering the relations between mechanical and molecular/cellular signatures in health and disease. I will discuss how blood vessel cells interact mechano-chemically with each other to regulate the passage of cells and nutrients between blood and tissue and how cancer cells grow and die in response to mechanical and geometrical stimuli.

Fri, 27 May 2022

14:00 - 15:00
L6

Coping with mechanical stress: tissue dynamics in development and repair

Prof Yanlan Mao
(Laboratory for Molecular Cell Biology UCL)
Abstract

During growth and development, tissue dynamics, such as tissue folding, cell intercalations and oriented cell divisions, are critical for shaping tissues and organs. However, less is known about how tissues regulate their dynamics during tissue homeostasis and repair, to maintain their shape after development. In this talk, we will discuss how differential growth rates can generate precise folds in tissues. We will also discuss how tissues respond to mechanical perturbations, such as stretching or wounding, by altering their actomyosin contractile structures, to change tissue dynamics, and thus preserve tissue shape and patterning. We combine genetics, biophysics and computational modelling to study these processes.

Fri, 20 May 2022

14:00 - 15:00
L6

Causal inference, big data and public health: estimating effectiveness and quantifying waning effectiveness of COVID-19 vaccines

Prof Jonathan Sterne
(Department of Population Health Sciences University of Bristol)
Abstract

Effectiveness of COVID-19 vaccines was first demonstrated in randomised trials, but many questions of vital importance to vaccination policies could only be addressed in subsequent observational studies. The pandemic led to a step change in the availability of population-level linked electronic health record data, analysed in privacy-protecting Trusted Research Environments, across the UK. I will discuss methodological approaches to estimating causal effects of COVID-19 vaccines, and their application in estimating vaccine effectiveness and quantifying waning vaccine effectiveness. I will present results from recent analyses using detailed linked data on up to 24 million people in the OpenSAFELY Trusted Research Environment, which was developed by the University of Oxford's Bennett Institute for Applied Data Science.

Fri, 13 May 2022

14:00 - 15:00
L6

Integrative analytics connecting genotype and phenotype for precision oncology

Dr Ian Overton
(School of Medicine Dentistry and Biomedical Science Queens University Belfast)
Abstract

Understanding the molecular mechanisms that control the biology of health and disease requires development of models that traverse multiple scales of organisation in order to encapsulate the relationships between genes and linking to observable phenotypes. Measuring, parameterising and simulating the entire system that determines these phenotypes in exhaustive detail is typically impossible due to the underlying biological complexity, our limited knowledge and the paucity of available data. For example, approximately one third of human genes are poorly characterised and most genes perform multiple functions, which manifest according to the surrounding biochemical context. Indeed, new functions continue to emerge even for deeply studied genes. Therefore, simplifying abstractions in concert with empirical analysis of matched genome-scale and descriptive data are valuable strategies to fill knowledge gaps relevant to a focused biomedical question or hypothesis.

Epithelial plasticity is a key driver of cancer progression and is associated with the most life-threatening phenotypes; specifically, metastasis and drug resistance. Computational methods developed in my group enable modelling the molecular control of important cancer phenotypesWe applied a machine learning approach for genome-wide context-specific biochemical interaction network inference (CoSNI) to map gene function for the Epithelial to Mesenchymal Transition cell programme (EMT_MAP), predicting new mechanisms in control of cancer invasion. Analysis of patient data with EMT_MAP and our NetNC algorithm [Cancers 2020;12:2823; https://github.com/overton-group/NetNC] enabled discovery of candidate renal cancer prognostic markers with clear advantages over standard statistical approaches. NetNC recovers the network-defined signal in noisy data, for example distinguishing functional EMT Transcription Factor targets from ‘neutral’ binding sites and defining biologically coherent modules in renal cancer drug response time course data. These and other approaches, including SynLeGG (Nucleic Acids Research 2021;49:W613-8, www.overton-lab.uk/synleggand an information-theoretic approach to causality (GABIoffer mechanistic insights and opportunity to predict candidate cancer Achilles’ heels for drug discovery. Computational results were validated in follow-up experiments, towards new clinical tools for precision oncology.

Fri, 06 May 2022

14:00 - 15:00
L6

Intrinsic instability of the dysbiotic microbiome revealed through dynamical systems inference at ecosystem-scale

Dr Travis Gibson
(Harvard Medical School)
Abstract

Dynamical systems models are a powerful tool for analyzing interactions in ecosystems and their intrinsic properties such as stability and resilience. The human intestinal microbiome is a complex ecosystem of hundreds of microbial species, critical to our health, and when in a disrupted state termed dysbiosis, is involved in a variety of diseases.  Although dysbiosis remains incompletely understood, it is not caused by single pathogens, but instead involves broader disruptions to the microbial ecosystem.  Dynamical systems models would thus seem a natural approach for analyzing dysbiosis, but have been hampered by the scale of the human gut microbiome, which constitutes hundreds of thousands of potential ecological interactions, and is profiled using sparse and noisy measurements. Here we introduce a combined experimental and statistical machine learning approach that overcomes these challenges to provide the first comprehensive and predictive model of microbial dynamics at ecosystem-scale. We show that dysbiosis is characterized by competitive cycles of interactions among microbial species, in contrast to the healthy microbiome, which is stabilized by chains of positive interactions initiated by resistant starch-degrading bacteria. To achieve these results, we created cohorts of “humanized” gnotobiotic mice via fecal transplantation from healthy and dysbiotic human donors, and subjected mice to dietary and antibiotic perturbations, in the densest temporal interventional study to date. We demonstrate that our probabilistic machine learning method achieves scalability while maintaining interpretability on these data, by inferring a small number of modules of bacterial taxa that share common interactions and responses to perturbations. Our findings provide new insights into the mechanisms of microbial dysbiosis, have potential implications for therapies to restore the microbiome to treat disease, and moreover offer a powerful framework for analyzing other complex ecosystems.

Fri, 29 Apr 2022

14:00 - 15:00
Virtual

Spreading mechanics and differentiation of astrocytes during retinal development

Prof Tracy Stepien
(Department of Mathematics University of Florida)
Abstract

In embryonic development, formation of the retinal vasculature is  critically dependent on prior establishment of a mesh of astrocytes.  
Astrocytes emerge from the optic nerve head and then migrate over the retinal surface in a radially symmetric manner and mature through 
differentiation.  We develop a PDE model describing the migration and  differentiation of astrocytes, and numerical simulations are compared to 
experimental data to assist in elucidating the mechanisms responsible for the distribution of astrocytes via parameter analysis. This is joint 
work with Timothy Secomb.

Fri, 11 Mar 2022

14:00 - 15:00
L3

Examples of artificial intelligence uses in target identification and drug discovery

Dr Ramneek Gupta
(Novo Nordisk Research Centre Oxford University of Oxford)
Abstract

As biological data has become more accessible and available in biology and healthcare, we find increasing opportunities to leverage artificial intelligence to help with data integration and picking out patterns in complex data. In this short talk, we will provide glimpses of what we do at the Novo Nordisk Research Centre Oxford towards understanding patient journeys, and in the use of knowledge graphs to draw insights from diverse biomedical data streams