Forthcoming events in this series


Fri, 07 Feb 2020

14:00 - 15:00
L3

Systems biology for single cell RNA-Seq data

Dr Tom Thorne
(Dept of Computer Science University of Reading)
Abstract

Single cell RNA-Seq data is challenging to analyse due to problems like dropout and cell type identification. We present a novel clustering 
approach that applies mixture models to learn interpretable clusters from RNA-Seq data, and demonstrate how it can be applied to publicly 
available scRNA-Seq data from the mouse brain. Having inferred groupings of the cells, we can then attempt to learn networks from the data. These 
approaches are widely applicable to single cell RNA-Seq datasets where  there is a need to identify and characterise sub-populations of cells.

 

Fri, 24 Jan 2020

14:00 - 15:00
L3

Mathematical modelling as part of an HIV clinical trial in sub-Saharan Africa

Dr Will Probert
(Big Data Institute Nuffield Department of Medicine University of Oxford)
Abstract

Globally, almost 38 million people are living with HIV.  HPTN 071 (PopART) is the largest HIV prevention trial to date, taking place in 21 communities in Zambia and South Africa with a combined population of more than 1 million people.  As part of the trial an individual-based mathematical model was developed to help in planning the trial, to help interpret the results of the trial, and to make projections both into the future and to areas where the trial did not take place. In this talk I will outline the individual-based mathematical model used in the trial, the inference framework, and will discuss examples of how the results from the model have been used to help inform policy decisions.  

Fri, 29 Nov 2019

14:00 - 15:00
L3

Fluid mediated mechanical effects in biology of single cells: Hydrodynamics in strategies for early stage biofilm formation and DNA damage during migration in cancer cells

Dr Rachel Bennett
(School of Mathematics University of Bristol)
Abstract

In the first part of the talk, I will describe surface colonization strategies of the motile bacteria Pseudomonas aeruginosa. During early stages of biofilm formation, the majority of cells that land on a surface eventually detach. After a prolonged lag time, cells begin to cover the surface rapidly. Reversible attachments provide cells and their descendants with multigenerational memory of the surface that primes the planktonic population for colonization. Two different strains use different surface sensing machinery and show different colonization strategies. We use theoretical modelling to investigate how the hydrodynamics of type IV pili and flagella activity lead to increased detachment rates and show that the contribution from this hydrodynamic effect plays a role in the different colonization strategies observed in the two strains.

In the second part of the talk, I will show that when cells migrate through constricting pores, there is an increase in DNA damage and mutations. Experimental observations show that this breakage is not due to mechanical stress. I present an elastic-fluid model of the cell nucleus, coupled to kinetics of DNA breakage and repair proposing a mechanism by which nuclear deformation can lead to DNA damage. I show that segregation of soluble repair factors from the chromatin during migration leads to a decrease in the repair rate and an accumulation of damage that is sufficient to account for the extent of DNA damage observed experimentally.

Fri, 22 Nov 2019

14:00 - 15:00
L3

Uncovering the mechanisms of mutagenesis: from dry lab to wet lab and back again

Miss Marketa Tomkova
(Nuffield Dept of Medicine University of Oxford)
Abstract

Understanding the mechanisms of mutagenesis is important for prevention and treatment of numerous diseases, most prominently cancer. Large sequencing datasets revealed a substantial number of mutational processes in recent years, many of which are poorly understood or of completely unknown aetiology. These mutational processes leave characteristic sequence patterns in the DNA, often called "mutational signatures". We use bioinformatics methods to characterise the mutational signatures with respect to different genomic features and processes in order to unravel the aetiology and mechanisms of mutagenesis. 

In this talk, I will present our results on how mutational processes might be modulated by DNA replication. We developed a linear-algebra-based method to quantify the magnitude of replication strand asymmetry of mutational signatures in individual patients, followed by detection of these signatures in early and late replicating regions. Our analysis shows that a surprisingly high proportion (more than 75 %) of mutational signatures exhibits a significant replication strand asymmetry or correlation with replication timing. However, distinct groups of signatures have distinct replication-associated properties, capturing differences in DNA repair related to replication, and how different types of DNA damage are translated into mutations during replication. These findings shed new light on the aetiology of several common but poorly explained mutational signatures, such as suggesting a novel role of replication in the mutagenesis due to 5-methylcytosine (signature 1), or supporting involvement of oxidative damage in the aetiology of a signature characteristic for oesophageal cancers (signature 17). I will conclude with our ongoing work of wet-lab validations of some of these hypotheses and usage of computational methods (such as genetic algorithms) in guiding the development of experimental protocols.

Fri, 15 Nov 2019

14:00 - 15:00
L3

Emergent spatial patterning in engineered bacteria

Dr Neil Dalchau
(Microsoft Research Cambridge)
Abstract

The spatial coordination of cellular differentiation enables functional organogenesis. How coordination results in specific patterns of differentiation in a robust manner is a fundamental question for all developmental systems in biology. Theoreticians such as Turing and Wolpert have proposed the importance of specific mechanisms that enable certain types of patterns to emerge, but these mechanisms are often difficult to identify in natural systems. Therefore, we have started using synthetic biology to ask whether specific mechanisms of pattern formation can be engineered into a simple cellular background. In this talk, I will show several examples of emergent spatial patterning that results from the insertion of synthetic signalling pathways and transcriptional logic into E. coli. In all cases, we use computational modelling to initially design circuits with a desired outcome, and improve the selection of biological components (DNA sub-sequences) that achieve this outcome according to a quantifiable measure. In the specific case of Turing patterns, we have yet to produce a functional system in vivo, but I will describe new analytical tools that are helping to guide the design of synthetic circuits that can produce a Turing instability.

Fri, 25 Oct 2019

14:00 - 15:00
L3

Embryogenesis: a cascade of dynamical systems

Professor Stanislav Shavrtsman
((Dept of Physical and Biological Engineering Princeton University)
Abstract

We aim to establish and experimentally test mathematical models of embryogenesis. While the foundation of this research is based on models of isolated developmental events, the ultimate challenge is to formulate and understand dynamical systems encompassing multiple stages of development and multiple levels of regulation. These range from specific chemical reactions in single cells to coordinated dynamics of multiple cells during morphogenesis. Examples of our dynamical systems models of embryogenesis – from the events in the Drosophila egg to the early stages of gastrulation – will be presented. Each of these will demonstrate what had been learned from model analysis and model-driven experiments, and what further research directions are guided by these models.

Fri, 18 Oct 2019

14:00 - 15:00
L3

Cell polarity formation and the dynamics of small G proteins; or, why your Turing bifurcations should always be subcritical

Professor Alan Champneys
(Dept of Engineering Maths University of Bristol)
Abstract

In this talk I shall describe recent work inspired by problems in cell biology, namely how the dynamics of small G-proteins underlies polarity formation. Their dynamics is such that their active membrane bound form diffuses more slowly. Hence you might expect Turing patterns. Yet how do cells form backs and fronts or single isolated patches. In understanding these questions we shall show that the key is to identify the parameter region where Turing bifurcations are sub-critical. What emerges is a unified 2-parameter bifurcation diagram containing pinned fronts, localised spots, localised patterns. This diagram appears in many canonical models such as Schnakenberg and Brusselator, as well as biologically more realistic systems. A link is also found between theories of semi-string interaction asymptotics and so-called homoclinic snaking. I will close with some remarks about relevance to root hair formation and to the importance of subcriticality in biology. 

Fri, 21 Jun 2019

14:00 - 15:00
L2

Personalised predictive modelling for transcatheter mitral valve replacement

Dr Adelaide De Vecchi
(Department of Biomedical Engineering King’s College London)
Abstract

Mitral regurgitation is one of the most common valve diseases in the UK and contributes to 50% of the transcatheter mitral valve replacement (TMVR) procedures with bioprosthetic valves. TMVR is generally performed in frailer, older patients unlikely to tolerate open-heart surgery or further interventions. One of the side effects of implanting a bioprosthetic valve is a condition known as left ventricular outflow obstruction, whereby the implanted device can partially obstruct the outflow of blood from the left ventricle causing high flow resistance. The ventricle has then to pump more vigorously to provide adequate blood supply to the circulatory system and becomes hypertrophic. This ultimately results in poor contractility and heart failure.
We developed personalised image-based models to characterise the complex relationship between anatomy, blood flow, and ventricular function both before and after TMVR. The model prediction provides key information to match individual patient and device size, such as postoperative changes in intraventricular pressure gradients and blood residence time. Our pilot data from a cohort of 7 TMVR patients identified a correlation between the degree of outflow obstruction and the deterioration of ventricular function: when approximately one third of the outflow was obstructed as a result of the device implantation, significant increases in the flow resistance and the average time spent by the blood inside the ventricle were observed, which are in turn associated with hypertrophic ventricular remodelling and blood stagnation, respectively. Currently, preprocedural planning for TMVR relies largely on anecdotal experience and standard anatomical evaluations. The haemodynamic knowledge derived from the models has the potential to enhance significantly pre procedural planning and, in the long term, help develop a personalised risk scoring system specifically designed for TMVR patients.
 

Fri, 14 Jun 2019

14:00 - 15:00
L2

Reactions, diffusion and volume exclusion in a heterogeneous system of interacting particles

Dr Maria Bruna
(Mathematical Institute University of Oxford)
Abstract


Cellular migration can be affected by short-range interactions between cells such as volume exclusion, long-range forces such as chemotaxis, or reactions such as phenotypic switching. In this talk I will discuss how to incorporate these processes into a discrete or continuum modelling frameworks. In particular, we consider a system with two types of diffusing hard spheres that can react (switch type) upon colliding. We use the method of matched asymptotic expansions to obtain a systematic model reduction, consisting of a nonlinear reaction-diffusion system of equations. Finally, we demonstrate how this approach can be used to study the effects of excluded volume on cellular chemotaxis. This is joint work with Dan Wilson and Helen Byrne.
 

Fri, 07 Jun 2019

14:00 - 15:00
L3

Mechanobiology of cell migration: mathematical modelling and microfluidics-based experiments go hand-in-hand

Dr Jose Manuel Garcia Aznar
(Dept of Mechanical Engineering University of Zaragoza)
Abstract

Mechanobiology is a field of science that aims to understand how mechanics regulate biology. It focuses on how mechanical forces and alterations in mechanical properties of cell or tissues regulate biological processes in development, physiology and disease. In fact, all these processes occur in our body, which presents a clear structural and hierarchical organization that goes from the organism to the cellular level. To advance in the understanding of all these processes at different scales requires the use of simplified representations of our body, which is normally known as modelling or equivalently the creation of a model. Different types of models can be found in the literature: in-vitro, in-vivo and in-silico models.

Here, I will present our modelling strategy in which we integrate different mathematical models and experiments in order to tackle relevant mechanical-based mechanisms in wound healing and cancer metastasis progression [1,2]. In fact, we have focused our research on individual [3] and collective cell migration [4], because it is a crucial event in all these mechanisms. Therefore, unravelling the intrinsic mechanisms that cells use to define their migration is an essential element for advancing the development of new technologies in regenerative medicine and cancer.

Due to the complexity of all these mechanisms, mathematical modelling is a relevant tool for providing deeper insight and quantitative predictions of the mechanical interplay between cells and extracellular matrix during cell migration. To assess the predictive capacity of these models, we will compare our numerical results with microfluidic-based experiments [2], which provide experimental information to test and refine the main assumptions of our models.

Actually, we design and fabricate multi-channel 3D microfluidics cell culture chips, which allow recreating the physiology and disease of one organ or any biological process with a precise control of the micro environmental factors [5]. Therefore, this kind of organ-on-a-chip experiments constitutes a novel modelling strategy of in vitro multicellular human systems that in combination with mathematical simulations provide a relevant tool for research in mechanobiology.

References

Escribano J, Chen M, Moeendarbary E, Cao X, Shenoy V, Garcia-Aznar JM, Kamm RD, Spill F.  Balance of Mechanical Forces Drives Endothelial Gap Formation and May Facilitate Cancer and Immune-Cell Extravasation. PLOS Computational Biology, in press.

Fri, 31 May 2019

14:00 - 15:00
L3

Algorithmic generation of physiologically realistic patterns of fibrosis in the heart

Professor Kevin Burrage
(School of Mathematical Sciences Queensland University of Technology Brisbane)
Abstract

Cardiac fibrosis plays a significant role in the disruption of healthy electrical signalling in the heart, creating structural heterogeneities that induce and stabilise arrhythmia.  However, a proper understanding of the consequences of cardiac fibrosis must take into account the complex and highly variable patterns of its spatial localisation in the heart, which significantly affects the extent and manner of its impacts on cardiac wave propagation. In this work we present a methodology for the algorithmic generation of fibrotic patterns via Perlin noise, a technique for computationally efficient generation of textures in computer graphics.

Our approach works directly from image data to create populations of pattern realisations that all resemble the target image under a set of metrics. Our technique thus serves as a type of data enrichment, enabling analysis of how variability in the precise placement of fibrotic structures modulates their electrophysiological impact. We demonstrate our method, and the types of analysis it can enable, using a widely referenced histological image of four different types of microfibrotic structure. Our generator and Bayesian tuning method prove flexible enough to successfully capture each of these very distinct patterns.

We demonstrate the importance of this tool, by presenting 2D simulations overlayed on the generated images that highlight the effects of microscopic variability on the electrophysiological impact of fibrosis. Finally, we discuss the application of our methodology to the increasingly available imaging data of fibrotic patterning on a more macroscopic scale, and indeed to other areas of science underpinned by image based modelling and simulation.    

Fri, 17 May 2019

14:00 - 15:00
L3

Combining computational modelling, structural biology and immunology to understand Antigen processing

Professor Tim Elliott
(Dept of Medicine University of Southampton)
Abstract

Competition between peptides for binding and presentation by MHC class I molecules decides the immune response to foreign or tumor antigens. Many previous studies have attempted to classify the immunogenicity of a peptide using machine learning algorithms to predict the affinity, or half-life, of the peptide binding to MHC. However immunopeptidome analyses have shown a poor correlation between sequence based predictions and the abundance on the cell surface of the experimentally identified peptides. Such metrics are, for instance, only comparable when the abundance of competing peptides can be accurately quantified. We have developed a model for predicting the relative presentation of competing peptides that takes into account off-rate, source protein abundance and turnover and cofactor-assisted MHC assembly with peptides. This model is mechanism based so that it can accommodate complex biology phenomena such as inflammation, up or downregulation of peptide loading complex chaperones, appearance of a mutanome. We have used aspects of the model to drive an investigation of the precise molecular mechanism of peptide selection by MHC I and its associated intracellular cofactors.

Fri, 10 May 2019
00:00

None

PLEASE NOTE THAT THIS SEMINAR IS CANCELLED DUE TO UNFORESEEN CIRCUMSTANCES
Abstract

PLEASE NOTE THAT THIS SEMINAR IS CANCELLED DUE TO UNFORSEEN CIRCUMSTANCES.

Fri, 03 May 2019

14:00 - 15:00
L3

Biomechanics can provide a new perspective on microbiology

Professor Takuji Ishikawa
(Dept. Finemechanics Grad. Sch. Eng Tohoku University)
Abstract

Despite their tiny size, microorganisms play a huge role in many biological, medical, and engineering phenomena. For example, massive plankton blooms are an integral part of the oceanic ecosystem. Algal cells incorporate carbon dioxide, which affects global warming. In industry, microorganisms are used in bioreactors to produce food and medicines and to treat sewage. The human body hosts hundreds of microorganism species, and the number of microorganisms in the human body is roughly double the number of cells in the body. In the intestine, approximately 1 kg of enterobacteria form a unique ecosystem, called the gut flora, which plays important roles in digestion and in relation to infection. Because of the considerable influence that microorganisms have on human life, the study of their behavior and function is important.

Recent research has demonstrated the importance of biomechanics in understanding the behavior and functions of microorganisms. For example, red tides can be induced by the interplay between the background flow and swimming cells. A dense suspension of bacteria can generate a coherent structure, which strongly enhances mass transport in a suspension. These phenomena show that the physical environments around cells alter their behavior and biological functions. Such a biomechanical understanding is still lacking in microbiology, and we believe that biomechanics can provide new perspectives on future microbiology.

In this talk, we first introduce some of our studies of the behavior of individual swimming microorganisms near surfaces. We show that hydrodynamic forces can trap cells at liquid–air or liquid–solid interfaces. We then introduce interactions between a pair of swimming microorganisms, because a two-body interaction is the simplest many-body interaction. We show that our mathematical models can describe the interactions between two nearby swimming microorganisms. Collective motions formed by a group of swimming microorganisms are also introduced. We show that some collective motions of microorganisms, such as coherent structures of bacterial suspensions, can be understood in terms of fluid mechanics. We then discuss how cellular-level phenomena can change the rheological and diffusion properties of a suspension. The macroscopic properties of a suspension are strongly affected by mesoscale flow structures, which in turn are strongly affected by the interactions between cells. Hence, a bottom-up strategy, i.e., from a cellular level to a continuum suspension level, represents a natural approach to the study of a suspension of swimming microorganisms. Finally, we discuss whether our understanding of biological functions can be strengthened by the application of biomechanics, and how we can contribute to the future of microbiology.

Fri, 08 Mar 2019

14:00 - 15:00
L2

Arrhythmia from dyad to whole-heart: bi-directional coupling between re-entry and spontaneous calcium release

Dr Michael Colman
(Faculty of Biomedical Sciences University of Leeds)
Abstract

The mechanisms underlying the initiation and perpetuation of cardiac arrhythmias are inherently multi-scale: whereas arrhythmias are intrinsically tissue-level phenomena, they have a significant dependence cellular electrophysiological factors. Spontaneous sub-cellular calcium release events (SCRE), such as calcium waves, are a exemplars of the multi-scale nature of cardiac arrhythmias: stochastic dynamics at the nanometre-scale can influence tissue excitation  patterns at the centimetre scale, as triggered action potentials may elicit focal excitations. This latter mechanism has been long proposed to underlie, in particular, the initiation of rapid arrhythmias such as tachycardia and fibrillation, yet systematic analysis of this mechanism has yet to be fully explored. Moreover, potential bi-directional coupling has been seldom explored even in concept.

A major challenge of dissecting the role and importance of SCRE in cardiac arrhythmias is that of simultaneously exploring sub-cellular and tissue function experimentally. Computational modelling provides a potential approach to perform such analysis, but requires new techniques to be employed to practically simulate sub-cellular stochastic events in tissue-scale models comprising thousands or millions of coupled cells.

This presentation will outline the novel techniques developed to achieve this aim, and explore preliminary studies investigating the mechanisms and importance of SCRE in tissue-scale arrhythmia: How do independent, small-scale sub-cellular events overcome electrotonic load and manifest as a focal excitation? How can SCRE focal (and non-focal) dynamics lead to re-entrant excitation? How does long-term re-entrant excitation interact with SCRE to perpetuate and degenerate arrhythmia?

Fri, 22 Feb 2019

14:00 - 15:00
L3

Programming languages for molecular and genetic devices

Dr Andrew Phillips
(Head of Biological Computation Group Microsoft Research Cambridge)
Abstract

Computational nucleic acid devices show great potential for enabling a broad range of biotechnology applications, including smart probes for molecular biology research, in vitro assembly of complex compounds, high-precision in vitro disease diagnosis and, ultimately, computational therapeutics inside living cells. This diversity of applications is supported by a range of implementation strategies, including nucleic acid strand displacement, localisation to substrates, and the use of enzymes with polymerase, nickase and exonuclease functionality. However, existing computational design tools are unable to account for these different strategies in a unified manner. This talk presents a programming language that allows a broad range of computational nucleic acid systems to be designed and analysed. We also demonstrate how similar approaches can be incorporated into a programming language for designing genetic devices that are inserted into cells to reprogram their behaviour. The language is used to characterise the genetic components for programming populations of cells that communicate and self-organise into spatial patterns. More generally, we anticipate that languages and software for programming molecular and genetic devices will accelerate the development of future biotechnology applications.

Fri, 15 Feb 2019

14:00 - 15:00
L3

“How did that get there?” Modelling tissue age evolution of Barrett’s esophagus

Dr Kit Curtius
(Barts Cancer Institute Queen Mary University of London)
Abstract

There is great interest in the molecular characterisation of intestinal metaplasia, such as Barrett’s esophagus (BE), to understand the basic biology of metaplastic development from a tissue of origin. BE is asymptomatic, so it is not generally known how long a patient has lived with this precursor of esophageal adenocarcinoma (EAC) when initially diagnosed in the clinic. We previously constructed a BE clock model using patient-specific methylation data to estimate BE onset times using Bayesian inference techniques, and thus obtain the biological age of BE tissue (Curtius et al. 2016). We find such epigenetic drift to be widely evident in BE tissue (Luebeck et al. 2017) and the corresponding tissue ages show large inter-individual heterogeneity in two patient populations.               

From a basic biological mechanism standpoint, it is not fully understood how the Barrett’s tissue first forms in the human esophagus because this process is never observed in vivo, yet such information is critical to inform biomarkers of risk based on temporal features (e.g., growth rates, tissue age) reflecting the evolution toward cancer. We analysed multi-region samples from 17 BE patients to

1) measure the spatial heterogeneity in biological tissue ages, and 2) use these ages to calibrate mathematical models (agent-based and continuum) of the mechanisms for formation of the segment itself. Most importantly, we found that tissue must be regenerated nearer to the stomach, perhaps driven by wound healing caused by exposure to reflux, implying a gastric tissue of origin for the lesions observed in BE. Combining bioinformatics and mechanistic modelling allowed us to infer evolutionary processes that cannot be clinically observed and we believe there is great translational promise to develop such hybrid methods to better understand multiscale cancer data.

References:

Curtius K, Wong C, Hazelton WD, Kaz AM, Chak A, et al. (2016) A Molecular Clock Infers Heterogeneous Tissue Age Among Patients with Barrett's Esophagus. PLoS Comput Biol 12(5): e1004919

Luebeck EG, Curtius K, Hazelton WD, Made S, Yu M, et al. (2017) Identification of a key role of epigenetic drift in Barrett’s esophagus and esophageal adenocarcinoma. J Clin Epigenet 9:113

Fri, 08 Feb 2019

14:00 - 15:00
L3

Untangling heterogeneity in DNA replication with nanopore sequencing

Dr Michael Boemo
(Sir William Dunn School of Pathology University of Oxford)
Abstract

Genome replication is a stochastic process whereby each cell exhibits different patterns of origin activation and replication fork movement.  Despite this heterogeneity, replication is a remarkably stable process that works quickly and correctly over hundreds of thousands of iterations. Existing methods for measuring replication dynamics largely focus on how a population of cells behave on average, which precludes the detection of low probability errors that may have occurred in individual cells.  These errors can have a severe impact on genome integrity, yet existing single-molecule methods, such as DNA combing, are too costly, low-throughput, and low-resolution to effectively detect them.  We have created a method that uses Oxford Nanopore sequencing to create high-throughput genome-wide maps of DNA replication dynamics in single molecules.  I will discuss the informatics approach that our software uses, our use of mathematical modelling to explain the patterns that we observe, and questions in DNA replication and genome stability that our method is uniquely positioned to answer.