factories for advanced...

38
FACTORIES FOR ADVANCED BIOMANUFACTURING 6-7 TH DECEMBER 2017 HALIFAX HALL, SHEFFIELD

Upload: nguyentruc

Post on 21-Jul-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

FACTORIES FOR

ADVANCED

BIOMANUFACTURING 6-7TH DECEMBER 2017

HALIFAX HALL, SHEFFIELD

WELCOME

PROFESSOR JEFF GREEN, CBMNET DIRECTOR The imperative to revolutionise the chemicals industry by creating a sustainable bio-based future and the increasing importance of biologics in medicine pose major challenges to UK biotechnologists. The design and implementation of bespoke advanced microbial cell factories, that can reproducibly yield bio-based alternatives to the chemicals that underpin so much of modern infrastructure is a fundamental challenge. Chassis engineering represents the single most critical technology to revolutionise biomanufacturing by improving product yields, simplifying product recovery and improving sustainability through reduced materials use and waste, thereby enhancing process economics and commercial viability. With a strong research base already working on microbial chassis engineering, CBMNet is the natural progenitor for such an event centred on microbial chassis design.

EVENT AIM This event brings together over 130 academics with diverse interests in microbial chassis engineering, alongside industrial scientists who are interested in improving the robustness of whole-cell biocatalysts to overcome restrictions on product titres, separability, sustainability and process economics. Key themes for the event will address six major challenges in chassis design:

Reverse-engineering of microbial chassis, where the manufacturing process is designed first, and the biocatalyst is developed to match the process constraints

Tolerance to toxic products and substrates, to overcome current restrictions on product titres

Improved strain stability for continuous manufacturing, to achieve the productivity critical for bio-based bulk chemicals manufacturing

Improved atom efficiency, to enable efficient conversion of substrate to product without loss of carbon, by diverting fluxes away from cell growth towards product and minimizing by-product formation

Improved transport processes, to ensure efficient substrate uptake and product efflux

Innovative high throughput tools for systems metabolic engineering, including CRISPR-Cas, integrase technology, directed evolution, high-throughput screening and selection.

VENUE HALIFAX HALL HOTEL ENDCLIFFE VALE ROAD, SHEFFIELD S10 3ER

The conference and networking dinner all take place at Halifax Hall Hotel http://www.halifaxhall.co.uk/ Accommodation is at Halifax Hall (HH) and Jonas Apartments (JONAS) (see delegate table below for where your accommodation is)

GETTING THERE AIR The nearest airport is Manchester International Airport. From here, you can get to Sheffield on a direct train. https://www.thetrainline.com/ - search Manchester Airport to Sheffield. Alternatively, you can get a taxi from the Airport to the hotel.

TRAIN You can get a train to Sheffield from most UK cities. https://www.thetrainline.com/. From here, you can get to Halifax Hall by Bus or Taxi. There is a taxi rank at the station and Sheffield operates UBER https://get.uber.com/

BUS From the train station, take the number 120 bus from Sheffield bus station (Pond Street -Stop FS5). Dismount the bus at Endcliffe Crescent and Halifax Hall is then a 5-minute walk. From the University of Sheffield, take the number 120 bus from Glossop road, outside the 301 centre (S10 2HL). Dismount the bus at Endcliffe Crescent and Halifax Hall is then a 5-minute walk.

CAR There is parking on site at Halifax Hall for conference attendees for the duration of the event. Once you have parked, please ask reception staff for a permit to display in your windscreen.

AGENDA Time Title Person

Wednesday December 6th

10:00 Registration

10:15 Welcome Jeff Green, University of

Sheffield

10:30 Developing Disruptive Bio-Based Processes Adriana Botes, VideraBio

Session One: Improved Transport Processes

11:15 Understanding And Optimising Transport of Hemicellulose-Derived

Nutrients For Industrial Biotechnology & Bioenergy Gavin Thomas, University of

York

11:45 A Functional Model of The Gram-Positive Bacterial Signal Recognition

Particle And Potential Biotechnological Applications Colin Harwood, University of

Newcastle

12:05 Flash Presentation: Productive Bioconversion of Hydrophobic

Substrates Matthew Hodges, Oxford

Biotrans Limited

12:13 Flash Presentation: Engineering Glucan Polymer Uptake, Production

And Efflux Christian Voigt, University of

Sheffield

12.21 Lunch

Session Two: Product Tolerance

14:00 Multi-Omics Approaches For Understanding And Mitigating Product

Toxicity Dave Kelly, University of

Sheffield

14:30 Sustainable Production of Bio-N-Butanol: From Lab Bench To

Commercial Plant Holly Smith, Green Biologics

15:00 Flash Presentation: Microbial Resistance To Chlorhexidine Peter Henderson, University of

Leeds

15:10 Coffee

Session Three: Strain Stability

16:00 Genetic Stability For Continuous Chemicals Production In Bacterial

Fermentation Dr James Allen, UCL

16:30 New Applications In Industrial Microbiology A Driver For The

Development of New Genetic Control of Microbial Strains Phillipe Gabant, Syngulon

17:00 Strain Development And Spore Manufacture (For Microbiome

Applications) Daniela Heeg, CHAIN Biotech

17:10 Flash Presentation: Engineering Cupriavidus And Cyanobacteria For

Improved Production of High Value Chemicals Samantha Bryan, University of

Nottingham

17:18 Flash Presentation: Phosphite Utilisation As A Selective Marker In

Bacteria Andrew Hitchcock, University of

Sheffield

17:25 Flash Presentation: Improving lutein production in microalgae - a

genetic engineering approach Josie McQuillan, University of

Sheffield

17:32 Round Up Of The Day Jeff Green, University of

Sheffield

Session Four: Poster Presentations

18:30 Networking Drinks And Poster Session

1 Poster: A Model For The Functioning of The Gram-Positive Bacterial

Signal Recognition Particle Colin Harwood, Newcastle

University

2 Poster: Chassis Engineering For Monoterpenoid Production Gajendar Reddy, University of

Manchester

3 Poster: Metabolic Evolution of CHO Cells Alejandro Fernandez-Martell,

University of Sheffield

4 Poster: Metabolic Engineering of Saccharomyces Cerevisiae For Xylose

Fermentation For Bioethanol Production Farnaz Yusuf, ICGEB, New Delhi

5 Poster: Enzyme Engineering To Improve Ethylene Production In

Cupriavidus Necator Alexander Van Hagen,

University of Nottingham

6 Poster: Towards Photosynthetic Hydrogen Production Sean Craig, University of

Nottingham

8 Poster: Development of Genome Engineering Tools For Cupriavidus

Strains Sophie Vaud, University of

Nottingham

9 Poster: Engineering Cupriavidus Metallidurans Towards Acetaldehyde

Production Vera Salgado, University of

Nottingham

10 Poster: Potential of Extremophilic Enzymes In Biotechnology Roumiana Todorova, Bulgarian

Academy of Sciences

11 Poster: Metabolic Engineering To Improve Ethylene Production In

Cupriavidus Necator Raj Patel, University of

Nottingham

12 Poster: Impact of Microbial Transporters On Advanced Biofuels And

Biochemical Generations Dipankar Ghosh, ICL

13 Poster: Bio-Ethylene Production Using Plant Pathway In Cupriavidus

Spp. Pierre Reitzer, University of

Nottingham

14 Poster: Metabolic Engineering of Cupriavidus Necator H16 For

Production Of Platform Chemicals Katalin Kovacs, University of

Nottingham

15 Poster: The Genetic Basis of 3-Hydroxypropanoate Metabolism In

Cupriavidus Necator H16 Christian Arenas, University of Nottingham

16 Poster: Optimising Triterpenoid Production In Saccharomyces Cerevisiae Matthew Dale, University of

Edinburgh

17 Poster: Engineering Cupriavidus Necator For The Production Of 1,3-

Butanediol Using Synthetic Metabolic Pathways Joshua Gascoyne, University of

Nottingham

18 Poster: The Use of M. Psychrotolerans As An Alternative Chassis For Bio-

Manufacturing Nikolaos Pantidos, University of Edinburgh

19 Poster: Project DETOX: Detoxifying Bio-Based Production Joyce Bennett, University of

York

20

Poster: Structural Basis For High-Affinity Adipate Binding To Adpc 1

(RPA4515), An Orphan Periplasmic Binding Protein From The Tripartite

Tricarboxylate Transporter (TTT) Family In Rhodopseudomonas Palustris Leo Rosa, University of Sheffield

21

Poster: Understanding and controlling the phenotypic switch from a

yeast-like to hyphal phenotype in Candida tropicalis for use in Industrial

Biotechnology Steven Bourne, Aberystwyth University

22

Poster: Molecular tools to engineer cyanobacteria for industrial

biotechnology Mary Ann Madsen, University of Glasgow

23

Poster: Synthetic biology for electrosynthetic bioproduction by

Rhodopseudomonas palustris Robert Bradley, Imperial College London

19:30 Networking Dinner

Thursday December 7th

08:30 Welcome Gavin Thomas, University of

York

08:45 Next Generation Tools For Rapid Biomanufacturing Mike Lynch, Duke University,

North Carolina

Session Five: Atom Efficient Pathways

09:30 Negative Carbon In Bio-Based Substrate Conversions Into Sustainably

Produced Chemicals Philip Weyrauch, Ingenza

10.00 Machine-Learning Driven Analysis And Optimisation Of Metabolic

Networks Claudio Angione, University of

Teesside

10.20 Flash Presentation: Metabolic Engineering of Saccharomyces Cerevisiae

For Xylose Fermentation For Bioethanol Production Farnaz Yusuf, ICGEB, New Delhi

10.28 Flash Presentation: A Rapid Pathway Design And Optimisation Pipeline

For Chemical Production In Bacteria Adrian Jervis, University of

Manchester

10.36 Flash Presentation: Biotech Platform For Xylitol Production – A Tale Of

Metabolic Engineering And Intracellular Redox Balance Abhishek Somani, Aberystwyth

University

10.44 Coffee

Session Six: Bioprocess Design

11:15 Bioprocess Design: Bio-Based Methacrylates

Gill Stephens, University of Nottingham

11:45 Rapid Enzyme Discovery And Scale Up Stuart West, Biocatalysts

12.05 Microbial Production of Flavour And Fragrance Isoprenoids Georg Lentzen, Isobionics BV

12.25 Flash Presentation: Design of Mammalian Cell Factories By Direct

Evolution Alejandro Fernandez-Martell,

University of Sheffield

12.33 Flash Presentation: Developing Yeast Strains For Biofuel Production Naseem Guar, ICGEB, New Delhi

12.41 Flash Presentation: Engineering Bacteria For Isoprene Biosynthesis Naglis Malys, University of

Nottingham

12.49 Flash Presentation: Introducing Calysta Uk Tithira Wimalasena, Calysta UK

12.57 Lunch

Session Seven: Technologies For Rapid Cell Factory Engineering

13:45 Technologies For Rapid Cell Factory Engineering John Heap, ICL

14:15 Metabolic Engineering of Thermophiles And Genetic Tools Gudmundur Oli Hreggvidsson,

Matis

14:45 Flash Presentation: Serine Integrase Recombinational Assembly (SIRA)

For Rapid Multipart DNA Assembly And Rearrangement Femi Olorunniji, Liverpool John

Moores University

Session Eight: LCA, Techno-Economic Feasibility, RRI

15:00 Techno-Economic And Life Cycle Environmental Aspects Of Bio-Based

Products Jon McKechnie, University of

Nottingham

15:30 Mobilising Ideas of Responsible Innovation Susan Molyneux-Hodgson,

University of Exeter

15:50 Flash Presentation: Business Services For SMEs In The Bioeconomy:

Biobase4sme And Superbio Lucy Montgomery, NNFCC

16:00 Concluding Remarks 16:15 Close

SPEAKERS IN PRESENTING ORDER

Dr Adriana Botes Director and CSO, VideraBio [email protected] http://www.viderabio.com/ Research Interests: Translation of advances in synthetic biology & metabolic engineering for applications in Industrial Biotechnology: Design and construction of platform microbial cell factories and development of intensified bioprocesses to manufacture bioactive natural products. The use of platform microbial cell factories allows multiple products to be accessible via common metabolic precursors with incremental further metabolic engineering, thereby reducing development cost for additional products.

Next Generation Tools for Rapid Biomanufacturing In 2000, the US National academy of sciences identified Bio-based industrial products as a research and commercialisation priority and set out a vision for the future whereby the US would lead the global transition to bio-based industrial products. Bio-based fuels and chemicals are consistently flagged as one of the key technologies with significant potential to drive economic impact and disruption by 2025. Advances in enabling technologies during the last decade drove down the development cost of industrial microbial cell factories; yet there are few commercial successes to date. The obstacles faced in the commercialisation of bio-based chemicals and the lessons learned are discussed using selected case studies. The UK is usually heralded as an early adopter of innovation and has a strong research base in the development of the enabling technologies that underpins the development of industrial microbial cell factories. Despite this, the UK has a fledgling industrial biotechnology industry for the manufacturing of bio-based chemicals and materials relative to the US and other leading countries. The reasons for this discrepancy are briefly discussed.

Professor Gavin Thomas

Professor of Microbiology, University of York [email protected] http://thomaslabyork.weebly.com/ Research Interests: Bacterial transporters and membrane-bound enzymes, with applications in biotechnology and medicine.

Understanding and Optimising Transport of Hemicellulose-Derived Nutrients for Industrial Biotechnology & Bioenergy Releasing the full nutritional capabilities of lignocellulose, in the time-scales required for industrial biotechnology & bioenergy processes, is a major challenge. For hemicellulose, the ease of accessibility of the substrate is complicated by the heterogeneous nature of the glycan. While much is known about the enzymes required to deconstruct hemicellulose, the complementary suite of transporters that are used to assimilate the released sugars are poorly studied in important lignocellulose degraders. We describe our work in the discovery of new transporters specifically adapted for growth on hemicellulose including novel transporters for arabinofuranose and xylo-oligomers and the potential for transporter engineering as a tool for improving microbial cell factories.

Professor Colin Harwood Professor of Molecular Microbiology, Newcastle University [email protected] http://www.ncl.ac.uk/cbcb/staff/profile/colinharwood.html Research Interests: Development of tools for refactoring of members of the genus Bacillus using synthetic biology approaches and a range of molecular biological tools. Fundamental and applied aspects of protein secretion in members of the genus Bacillus.

A Functional Model of the Gram-Positive Bacterial Signal Recognition Particle and Potential Biotechnological Applications There is a great deal of uncertainty about the intracellular events associated with bacteria protein secretion. In E. coli the three key intracellular players are the Signal Recognition Particle (SRP – a ribonucleoprotein complex of 4.5S RNA and Ffh), SecA and SecB. It is now generally accepted that the SRP is required for the secretion of integral membrane proteins, while the SecA pathway (with or without SecB) is required for proteins that are targeted beyond the cytoplasmic membrane. SRP and SecA/B both function to maintain their preprotein substrates in the secretion competent (essentially unfolded) state required for translocation through the narrow pore of the Sec translocase. In Gram-positive species SecB is absent and the RNA component of its SRP (scRNA, 271 nt) is significantly longer than of its E. coli counterpart (114 nt). Consequently, unlike E. coli, It forms an Alu domain, responsible in higher organisms for translational arrest and subsequent co-translational translocation. The presence of Alu domains in Eukaryal, Archael and Bacillus SRP RNA, but not E. coli, raises fundamental questions about how the SRP functions in bacteria. In particular, do Gram-positive and Gram-negative bacteria translocate their cargo co-translationally and, if so, do they use similar mechanisms? To address this question we have undertaken a detailed analysis of the composition of the Bacillus SRP, using a combination of bacterial-2 hybrid (BACTH), electromobility shift (EMSA) and microscale thermophoresis (MST) assays. We have also shown that a recently identified protein component of the SRP interacts with a key protein involved in ribosomal function, and provides an attractive model for triggering translational arrest. We will discuss the potential biotechnological applications of this model.

Dr Matthew Hodges Director of Commercial Operations, Oxford Biotrans Limited [email protected] http://oxfordbiotrans.com/ Research Interests: Biocatalysis, oxidation, fermentation, DSP

Productive Bioconversion of Hydrophobic Substrates Enzymes are highly desirable catalysts due to their potential for high specificity, selectivity and activity, within a largely aqueous (low impact) environment. However, challenges ensue when boundary conditions are suboptimal, as in the case of hydrophobic substrates, and these are compounded by the high titres needed for a commercially viable process. Many options are available to mitigate these issues, ranging from cell free systems, the use of biphasic processes, through to synbio solutions such as transporters and synthesis redesign. These will be discussed in the context of the commercialisation of Oxford Biotrans nootkatone process.

Miss Aritha Dornau PhD Student, University of York [email protected] https://www.linkedin.com/in/aritha-dornau-b6331669/ Research Interests: I am a microbiologist with a primary interest in biomanufacturing and bioprocess engineering using waste feedstocks. My PhD project aims to investigate the potential of organic municipal solid waste (OMSW) as a feedstock for producing renewable fuels and chemicals.

Rubbish to Resource: Exploring Municipal Solid Waste as a Feedstock for Biomanufacturing Population growth, urbanisation and increasing economic prosperity are escalating the rate of municipal solid waste (MSW) production. Globally, the majority of MSW ends up in landfills or is incinerated, polluting the environment and contributing to climate change. On average 40% of MSW is biodegradable and primarily of plant origin, making it an abundant and renewable source of lignocellulose for biorefinery applications. This BBSRC industrial-CASE project aims to investigate the potential of the organic fraction of MSW (OMSW) as a renewable feedstock for biofuel production. We work with an organic fibre derived from autoclave pre-treated MSW, produced by our industrial partner Wilson Bio-Chemical (www.wilsonbio-chemical.co.uk). The Wilson Fibre® contains about 42% polysaccharides, comprising ~38% cellulose and ~4% hemicellulose. Marker inhibitors including furfural, 5-hydroxymethyl furfural, levulinate, formate and vanillin are present at low levels compared to other feedstocks reported in the literature, but a wide range of metals are present which are potentially inhibitory to microbial growth. We have produced high-sugar hydrolysates from the MSW fibre by enzymatic hydrolysis with the industrial enzyme cocktail Cellic Ctec3 (Novozymes) and demonstrated that ethanologenic Escherichia coli LW06 is able to reach high cell densities on nutrient-supplemented MSW fibre hydrolysate. Future work will focus on further exploring the physiology of E. coli LW06 growing on MSW fibre hydrolysate and evaluating fermentation performance alongside a collection of diverse microbial species in order to identify promising strains for industrial biomanufacturing.

Dr Christian Voigt Lecturer, University of Sheffield [email protected] https://www.sheffield.ac.uk/aps/staff-and-students/acadstaff/voigt Research Interests: Molecular mechanisms in biopolymer synthesis and their adaption to biotechnological applications. In particular, I have a focus on the (1,3)-ß-glucan which is a cell wall component in fungi, involved in plant defence responses and used for carbohydrate storage in brown macroalgae. Based on its biochemical characteristics and biological function, optimising and engineering (1,3)-ß-glucan biosynthesis opens many options in increased plant defence, improved 2nd generation biofuel production as well as pharmaceutical applications where new production pathways can lead to more productive and efficient impact.

Engineering Glucan Polymer Uptake, Production and Efflux The natural biopolymer (1,3)-ß-glucan is a major component of the fungal cell wall, used for carbohydrate storage in

brown macroalgae and is involved in plant defence responses. In contrast to the (1,4)-ß-glucan cellulose, (1,3)-ß-glucan

is non-crystalline and easier to hydrolyse. Therefore, (1,3)-ß-glucan-enriched biomass is a novel source for 2nd

generation biofuel production. Adaption of yeast strains for enhanced uptake of (1,3)-ß-glucan hydrolysing products

by expression of a bacterial (1,3)-ß-glucan transporter facilitated elevated ethanol production from (1,3)-ß-glucan-rich

plant and marine biomass. Apart from its biotechnological potential in biofuel production, (1,3)-ß-glucan is a potent

immunostimulant associated with health promoting traits. Current (1,3)-ß-glucan production relies on purification

from yeast where additional cell wall components complicate and raise the cost for a highly purified end product.

Novel cell free and bacterial (1,3)-ß-glucan production systems can overcome this production barrier and facilitate

improved purification as well as opportunities for targeted modification to increase application value.

Professor Dave Kelly Chair of Microbial Physiology, University of Sheffield [email protected] https://www.sheffield.ac.uk/mbb/staff/davekelly Research Interests: Solute transport systems, Bacterial physiology, Metabolomics Proteomics

Multi-omics approaches for understanding and mitigating product toxicity Toxicity is one of the most serious barriers to commercialization of products made by Industrial Biotechnology (IB) methods. Vanillin is a high value product due to its use as a flavouring compound and its production by biotransformation using bacteria is well established. However, as a phenolic aldehyde, toxicity is a key issue, although the mechanism of this is relatively poorly understood. We have used a high-resolution global proteomics approach to elucidate the responses of E. coli to vanillin. The results of the proteomics analyses were used to identify and further investigate potential protein and pathway targets that could be manipulated to improve vanillin tolerance. In addition, using an adaptive laboratory evolution (ALE) approach four vanillin-tolerant strains of E. coli were evolved and their genomes sequenced. Specific SNPs and other changes were identified that have provided additional novel insights into the toxicity of vanillin. A range of gene targets for engineering a more vanillin-tolerant strain of E. coli are suggested by our results, that can also inform toxicity mitigation for other similar industrially-relevant compounds. In this talk, I will also give an overview of our BBSRC IB catalyst project DETOX, a collaboration between academics at York, Nottingham, Sheffield, Cambridge and Exeter, and UK-based biotechnology companies Lucite, Green Biologics, Ingenza and the CPI. The DETOX project aims to provide a systematic analysis of how IB chemicals poison bacterial cells using multi-omics analyses and provide solutions by reverse engineering production chassis bacteria that can be used by UK IB businesses.

Dr Holly Smith

Head of Fermentation, Green Biologics Limited [email protected] www.greenbiologics.com Research Interests: Solventogenic Clostridia, fermentation, renewable chemicals, strain engineering, adaptive laboratory evolution, product toxicity and tolerance.

Sustainable production of bio-n-butanol: From lab bench to commercial plant Green Biologics Ltd (GBL) is a renewable chemicals company based in Abingdon, UK with a wholly owned U.S. operating company, Green Biologics Inc. GBL has recently started up its first commercial production facility for renewable n-butanol and acetone in Little Falls, Minnesota. GBL’s high productivity Clostridium fermentation platform converts a wide range of sustainable feedstocks into high performance green chemicals, and through chemical synthesis, derivatives used by a growing global consumer and industrial products customer base. Challenges such as product toxicity and feedstock inhibitors can be overcome using a range of strain development techniques such as evolutionary engineering and targeted genome modification using GBL’s proprietary Crispr/Cas based genome editing technology, CLEAVE™. Work with CBMNet partners is focused on understanding the effects of n-butanol on the membrane and the mechanism of toxicity with a view to improving product tolerance.

Professor Peter Henderson Research Professor, University of Leeds [email protected] http://www.astbury.leeds.ac.uk/people/staff/staffpage.php?StaffID=PJFH Research Interests: Molecular mechanisms of membrane transport, Antibiotic resistance, Bioenergetics, Development of NMR and MS techniques to study membrane proteins, Enzyme kinetics and ligand binding studies

Microbial resistance to chlorhexidine Resistance of pathogenic microorganisms to antiseptics and antibiotics is becoming a serious threat to human and animal health. With colleagues in Australia we characterised a novel membrane protein, called AceI, responsible for the efflux of chlorhexidine from cells of Acinetobacter species, so conferring resistance to a widely-used antiseptic [1]. Genome analyses showed there are similar proteins in many proteobacteria, including pathogens, giving rise to a novel family of drug efflux proteins, designated the Proteobacterial Acinetobacter Chlorhexidine Efflux, ‘PACE’ family [1]. Genes encoding AceI homologues from 23 species of bacteria were transferred to the pTTQ18 plasmid vector, and transformed into Escherichia coli BL21(DE3) host cells, where the expression of each cloned gene in membrane fractions was identified in Coomassie stained SDS-PAGE gels and in Western blots detecting the His6-terminus of each protein. Out of all those investigated, seven genes were expressed at levels sufficient for production of proteins at a 30 litre fermentation scale. Each of these was then purified in mg quantities by IMAC. The integrity of the purified proteins was assessed by circular dichroism and by assaying binding to known or putative substrates. Several of the highly expressed AceI homologues conferred resistance to acriflavine, a nucleic acid intercalating biocide. We aim to establish whether common metabolites in bacteria are the natural substrates of PACE proteins. [1] Hassan et al. (2015) “Homologues of the Acinetobacter baumannii AceI transporter represent a new family of bacterial multidrug efflux systems”. mBio 6 (1) e01982-14, 1-5.

Dr James Allen PDRA, University College London [email protected] http://www.ucl.ac.uk/biochemeng/people/pdra/allen-j Research Interests: As part of the Innovate UK grant, ConBioChem, my research is focussed on biological production of commodity chemicals in continuous fermentation. With Prof. John Ward at UCL, I am focussed on developing the bacterial chassis and processes through which continuous production can be obtained.

Genetic stability for continuous chemicals production in bacterial fermentation The increasing instability of fossil fuel markets and international efforts to increase sustainable practises means that bacteria are rapidly becoming an attractive prospect as catalysts for the production of a huge number of products in varying fields. Unlike value-added goods and medicines, however, the economics of commodity chemical production have so far prevented a breakthrough towards bacterial-based processes. I describe these economic constraints and how they are beginning to change, along with which risks are still prominent in bacterial fermentation for chemicals production. Furthermore, I describe how far research has already come to demonstrating the extent of these risks and how the ConBioChem project is working to mitigate them, with particular emphasis on the generation of genetically stable strains to allow continuous fermentation processes.

Dr Philippe Gabant Chief scientific Officer, Syngulon [email protected] www.syngulon.com Research Interests: Syngulon is developing original genetic technologies to improve the efficiency of microorganisms (also called microrefineries) involved in industrial bioproduction. These new applications require deep genetic engineering (also called Synthetic Biology) of industrial microbial genomes to achieve high production yield and improve compatibility with the environment due to production in (semi-) open plants. The mission of Syngulon is to provide genetic technologies in order to make microbial strains involved in industrial process more efficient and safe

New applications in industrial microbiology a driver for the development of new genetic control of microbial strains Molecular biology techniques have, via synthetic biology, gained in sophistication and ease of use, leveraging the creativity associated to the definition of novel expression systems. In addition to industrial needs in terms of productivity and cost effectiveness, safety considerations became increasingly demanding. This is easely understood if we take into account the fact that in biobased production strains are used in non completely controlled environment. In industrial applications microbial strains need to be « protected » from surrounding microflora affecting negatively the process efficacy. The consequence was the definition or strengthening of new and logical quality and safety constraints standards progressively becoming mandatory and driving an impossible step backwards. Among the possible solution, we propose a general strategy using of bacteriocins that can provide an unique and innovative firewall to boost fermentation either in close or open environments. These natural effectors have recently been revisited for their potential applications and proven efficiency in bio-based production.

Dr Daniela Heeg Technical Product Manager, CHAIN Biotechnology Ltd [email protected] http://www.chainbiotech.com Research Interests: CHAIN Biotechnology Ltd develops spore forming Clostridium spp. for use as therapeutic delivery platforms. We are interested in manufacturing of Clostridium spp. for improved growth and spore yield and genetic engineering of those species for maximum product yield.

Strain development and spore manufacture (for microbiome applications) CHAIN Biotechnology Ltd is a SME developing novel microbiome based therapies for inflammatory and infectious diseases of the bowel. CADDTM has been developed as a platform to deliver novel therapeutics directly to affected areas. CADDTM is formulated using spores of Clostridium sp. that survive the adverse conditions in the upper intestinal tract, and subsequently germinate and grow in the lower intestinal tract to secrete the therapeutic substance. CHAIN has developed a genetically modified strain for secretion of its first product, a novel anti-inflammatory molecule. The spore product has been produced using fermentation and the spores have been validated for germination, growth and efficacy in in vitro pre-clinical tests. We plan to optimise the fermentation process for spore production and scale-up the fermentation and purification process to produce material to support in vivo pre-clinical studies. We will investigate growth conditions including mechanisms to induce sporulation. Here, we report on progress with these versatile anaerobic bacteria.

Samantha Bryan Senior Research Fellow, University of Nottingham [email protected] http://sbrc-nottingham.ac.uk/ Research Interests: Re-engineering C1 chassis for the bio production of high value compounds utilising metabolic engineering, synthetic biology and directed evolution. I am also extremely interested in developing HTP tools to accelerate metabolic engineering approaches including CRISPR and recombineering.

Engineering Cupriavidus and Cyanobacteria for Improved Production of High Value Chemicals Unlocking a sustainable, carbon neutral alternative to petrochemical production of high value chemicals is imperative. We aim to engineer both Cupriavidus sp. and Cyanobacteria as platforms for the production of ethylene, hydrogen and acetaldehyde. Ethylene is a small hydrocarbon gas, widely used in the chemical industry. Its annual worldwide production currently exceeds 150 million tonnes, surpassing any other organic compound. We aim to engineer Cupriavidus sp and Cyanobacteria as platforms for the production of ethylene utilising two different pathways, the ethylene forming enzyme (EFE) from P. syringae pv. Paseolicola and the Yang cycle from plants (ACO/ACS). Hydrogen is a promising alternative fuel; however traditional production methods release carbon and as such are unsustainable. Microbes such as the photosynthetic cyanobacteria produce hydrogen requiring only light, CO2 and water, and therefore offer the promise of clean, sustainable H2 production. We aim to develop a novel cyanobacterial chassis, underpinning a self-sustaining cellular factory to generate sustainable hydrogen. Acetaldehyde is an attractive C2 platform chemical, serving as a precursor for chemical catalytic conversion to both biofuel and biochemical products such as no-distil ethanol, 1-butanol and 1, 3-butadiene. Bio-production of aldehydes is gaining in interest yet presents challenges from both a toxicity and a by-product formation perspective. Preventing the reduction of aldehydes to alcohol by-products remains a metabolic engineering challenge. We aim to alleviate both the toxicity and by-product formation challenges associated with acetaldehyde bio-production.

Dr Andrew Hitchcock PDRA, University of Sheffield [email protected] https://www.sheffield.ac.uk/mbb/staff/postdocs/andrew_hitchcock Research Interests: Photosynthesis, cyanobacteria, photosynthetic bacteria, electron transport chains, pigment biosynthesis, nutrient uptake, synthetic biology, structural biology.

Phosphite utilisation as a selectable marker Phosphorus (P) is essential for life. It is mainly found as phosphate (PO43−), a component of DNA/RNA, nucleotide cofactors and phospholipids that is vital for cell metabolism, structure and signalling. In many natural environments the concentration of phosphate is so low that it is the growth-limiting nutrient. Microorganisms have therefore evolved the ability to acquire P from alternative sources, such as inorganic phosphite (HPO32-/HPO3H-). Phosphite is not only a source of P for some microorganisms and a potentially important part of the global P redox cycle, but as it is ecologically rare it has also been used in agriculture, industry and biotechnology, in conjunction with the phosphite dehydrogenase enzyme PtxD that oxidises it to phosphate, as a selectable marker, to provide competitive advantage, and as a biocontainment strategy. Natural phosphite utilisation requires an ABC-transporter for high affinity phosphite uptake in addition to the PtxD enzyme. In engineered systems, as phosphite is stable, water-soluble, cheap and non-toxic it can be provided in millimolar concentrations, which negates the requirement for the transporter as phosphite can be taken up non-specifically. However, the ability to use lower concentrations is desirable in some cases, and in nature the concentration of phosphite is orders of magnitude below that used in artificial systems, which necessitates high-affinity phosphite acquisition. We have determined the ligand binding properties and high-resolution X-ray crystal structures of the periplasmic binding protein components of four phosphite transporters. The potential to incorporate such transporters and lower the phosphite requirement for biotech applications will be discussed.

Miss Josie McQuillan PhD Student, University of Sheffield [email protected] https://www.sheffield.ac.uk/algae/researchers/josiemcquillan Research Interests: Developing and applying tools to genetically engineer microalgae (primarily Chlamydomonas) for the production of high-value terpenoid chemicals, and to further improve these organisms as chassis for industrial biotechnology. I am also interested in combining -omics techniques to identify and target areas of microalgal metabolism to manipulate with the aim of enhancing product yields.

Improving lutein production in microalgae - a genetic engineering approach Chlamydomonas reinhardtii is a fast-growing and genetically tractable model microalgal species with a wealth of genomic data, and capable of producing high-value products. Carotenoids, in particular lutein and zeaxanthin, are of value to several industries, and are naturally produced by this organism. Attempts to increase production of carotenoids in C. reinhardtii by overexpression of rate-limiting enzymes have resulted in small increases in carotenoid accumulation[1]. Identifying a relevant gene target is inherently difficult due to our limited understanding of the genetic basis of complex cellular processes. Here we identified and over-expressed an enzyme sourced from a plant able to post-translationally stabilise the carotenogenic enzyme phytoene desaturase, as well as by inducing chloroplast differentiation into carotenoid-rich chromophores. A homologue of the enzyme was identified within the C. reinhardtii genome via a BLAST search; this was cloned and overexpressed with the goal of increasing carotenoid, primarily lutein, production. This gene, whose introns were retained with the intention of enhancing recombinant gene expression[2], was inserted into a pOpt vector containing a paromomycin resistance cassette[2] downstream of constitutive hybrid promoter Hsp70A/RbcS2, as well as a poly-histidine tag; the vector was then transformed into a cell-wall deficient strain of C. reinhardtii via electroporation. Western blots and HPLC derived measurements of lutein and other pigments were undertaken on selected positive transformants, together with examination of chloroplast physiology in mutant strains that overexpress the GOI, using confocal microscopy. This project is a promising application of genetic engineering techniques to increase yields of high-value products in microalgal systems. [1] Couso et al. Biotechnol Prog 2011 27: 54 [2] Lauersen et al. Appl. Microbiol. Biotechnol. 2015 99: 3491

Dr Mike Lynch Assistant Professor Biomedical Engineering, Duke University, North Carolina [email protected] http://lynchlab.pratt.duke.edu/ Research Interests: Synthetic Biology, biochemical engineering, metabolic engineering, enzyme and protein engineering

Next Generation Tools for Rapid Biomanufacturing We report a high-throughput metabolic engineering platform enabling the rapid optimization of microbial production strains. The platform, which bridges a gap between current in vivo and in vitro bio-production approaches, relies on dynamic minimization of the active metabolic network. Dynamic metabolic network minimization is accomplished using combinations of CRISPR interference and controlled proteolysis to reduce the activity of multiple enzymes in essential central metabolism. Minimization is implemented in the context of standardized 2-stage bio-processes. This approach not only results in a design space with greatly reduced complexity, but also in increased metabolic fluxes and production rates as well as in strains which are robust to environmental conditions. Robustness leads to predictable scalability from high-throughput small-scale screens, or “micro-fermentations”, to fully instrumented bioreactors. Predictive high-throughput approaches are critical for metabolic engineering programs to truly take advantage of the rapidly increasing throughput and decreasing costs of synthetic biology. We have not only demonstrated proof of principle for this approach in two common industrial microbes: E. coli and S. cerevisiae, but also have validated this approach with the rapid optimization of E. coli strains producing numerous important chemicals.

Dr Philip Weyrauch Senior Biochemist, Ingenza Ltd [email protected] www.ingenza.com Research Interests: Industrial biotechnology, synthetic biology, efficient conversion of substrate to product without loss of carbon.

Negative carbon in bio-based substrate conversions into sustainably produced chemicals Succinate is widely recognised as a platform chemical (production at 30,000-50,000 tonne/annum) which has already received attention within the context of industrial biotechnology as a model for sustainable manufacturing. However, to permit current fermentative succinate production to co-utilise glucose and CO2 to maximise carbon efficiency, additional exogenous energy is required which can be supplied as a substrate. Here we propose to supply this additional energetic substrate in the form of renewable hydrogen gas. We have already obtained evidence that demonstrates efficient cellular processing of both CO2 and hydrogen gas to generate formate as part of a previous investigation. Furthermore, Ingenza has already demonstrated an efficient glucose to succinate fermentation process, which when supplemented with formate, can co-utilise CO2 as a tandem feedstock. Building on these important results we seek to deliver the cutting edge innovation of combining glucose, CO2 and renewable hydrogen to significantly enhance the efficiency of bioprocess carbon conversion beyond that possible with glucose and CO2 alone. We believe this approach is deployable on a wider scale to minimise and eliminate CO2 arising in biobased chemical manufacturing, where we are specifically seeking to demonstrate this capacity using succinate as a model system. It is envisaged this methodology could be adopted for more signficantly reduced products relative to succinate, providing an opportunity to increase CO2 abatement even further compared to conventional sugar/biomass derived fermentation processes.

Dr Claudio Angione Senior Lecturer, Teesside University [email protected] https://www.scm.tees.ac.uk/c.angione/#Research Interests: At the intersection of computer science, mathematics and biology. They include systems biology, metabolic engineering, genome-scale metabolic models, cancer metabolism, machine learning, multi-objective optimisation.

Machine-learning driven analysis and optimisation of metabolic networks Although most studies in biology mainly target genomics, transcriptomics and proteomics, genes and their expression alone do not always constitute a reliable indicator of cellular phenotype. However, the intrinsic complexity of cell metabolism - the dense network of biochemical reactions occurring in a cell - makes it difficult to predict the phenotypic traits resulting from a given genotype and a given environmental or disease condition. To this end, mathematical models and computational methods represent powerful abstraction tools for metabolic networks. In particular, genome-scale models of metabolism contain all known metabolic reactions in the cell, and therefore provide a snapshot that is closer to the observable phenotype. I will present a set of computational methods to create and investigate poly-omic models of bacterial and human metabolism. I will show how, by mapping omic data onto these models, it is possible to obtain an "enriched" view of any given gene or protein expression profile. This includes the predicted growth rate of a bacterial or human cell in any given condition, and flux rates for all its metabolic reactions. Finally, I will show that condition-, tissue- and patient-specific metabolic models shed light on cell-specific changes occurring in the metabolic network, therefore mechanistically predicting biomarkers for disease metabolism or cell engineering for biotechnological applications.

Dr Farnaz Yusuf National Postdoctoral Fellow, International Centre for Genetic Engineering and Biotechnology, New Delhi-India [email protected] https://www.icgeb.org/yeast-biofuel-group.html Research Interests: Metabolic engineering and synthetic biology, focused on understanding and improving the xylose metabolic pathway in S. cerevisiae for efficient and rapid utilization of xylose for bioethanol production. My research aims to construct a yeast strain for converting lignocellulosic biomass into sustainable fuels and developing this yeast into a more robust, inhibitor-tolerant cell factory.

Engineering Saccharomyces cerevisiae for xylose fermentation for biofuel production The depleting fossil fuel reserves, rising energy demands and the global climate change due to emission of greenhouse gases has switched the focus to the production of biofuels. Biofuels offer a promising cleanest renewable alternative source of energy to meet the energy consumption across the globe. Sustainable biofuel production from renewable biomass requires the efficient and complete use of all abundant sugars in the lignocellulosic biomass. Xylose is the main pentose and second most abundant sugar after glucose in lignocellulosic material. Saccharomyces cerevisiae lacks xylose metabolizing genes therefore, efforts are being made to engineer S. cerevisiae strains for efficient xylose fermentation to make the process economically feasible. The present study aims to construct a xylose fermenting yeast strains with engineered oxido-reductative pathway for xylose metabolism. The study also shows the effect of different promoters on xylose fermentation. The engineered strain is able to grow on xylose as sole carbon source with the maximum ethanol yield of 0.26g/g xylose and productivity of 0.07g/l/h at 96 hours. The further improvement in strain development involves over expression of pentose phosphate pathway and protein engineering of xylose reductase/xylitol dehydrogenase to change their cofactor specificity which is considered as one of the bottlenecks in efficient xylose fermentation.

Dr Adrian Jervis Senior Experimental Officer, University of Manchester [email protected] www.synbiochem.co.uk Research Interests: Developing synthetic biology technology for the microbial production of fine chemicals in the Manchester Centre for Synthetic Biology of Fine and Speciality Chemicals (SYNBIOCHEM). With a background in Molecular Microbiology I specialise in pathway design and assembly, chassis engineering and high-throughput assay development.

A rapid pathway design and optimisation pipeline for chemical production in bacteria The microbial production of fine chemicals provides a promising bio-sustainable manufacturing solution that could replace many present industrial chemical production and extraction processes. During the past decade, this approach has already led to the successful production of a growing catalogue of natural products and high-value chemicals in industrial microorganisms. Here we show that through the coordinated development and application of a suite of interdisciplinary tools, integration/automation of technology platforms and experimental protocols into a full Design-Build-Test-Learn pipeline strategy, the process can be accelerated. As a benchmark case, the pipeline was applied to the production of the flavonoid pinocembrin in Escherichia coli. The application of several rounds of combinatorial design resulted in optimized production strains for flavonoid compounds that could be further derivatized. By means of this strategy, microbial production of fine chemicals, a process that to date has proved to be resource intensive, becomes rationalized, flexible and increasingly efficient.

Dr Abhishek Somani PDRA, Aberystwyth University [email protected] https://www.aber.ac.uk/en/ibers/staff-profiles/listing/profile/abs12 Research Interests: Use of synthetic biology, metabolic engineering, fermentation sciences, systems biology and protein engineering to develop microbial chassis for metabolite production. Am also enthused by developing physicochemical methodologies for manipulating lignocellulosic biomass, scaling-up technologies for commercial applications and technoeconomic evaluations of the integrated biorefinery supply chain to develop innovative solutions for sustainable manufacturing.

Biotech Platform for Xylitol Production – A Tale of Metabolic Engineering and Intracellular Redox Balance To establish the environmental and economic sustainability of second generation lignocellulosic sugars as a biorefining platform, it is important to develop and optimise new biological routes to generate value added chemical commodities from low value hemicellulose sugars, predominantly xylose. With an estimated market of 1 Bn US$ by 2020, the natural sweetener xylitol is an attractive candidate to derive value from xylose. Despite considerable research into the bioconversion of xylose to xylitol using different microorganisms, commercial production of the sugar alcohol is still largely achieved via chemical synthesis. Using a new, inhibitor-tolerant Candida tropicalis isolate we employed strategies in traditional metabolic engineering along with rational, in silico enzyme re-design to attain intracellular redox balance and maximal xylitol yields from lignocellulose-derived xylose. Alongside the synthetic biology approach, the associated fermentation parameters were optimised (in both batch and fed-batch approaches) and xylose to xylitol bioconversion productivity was markedly enhanced (>2 fold) following process scale-up (150 L wheat straw hydrolysate fermentation). The cellular transcriptomic response was also evaluated, via RNA sequencing, and provided insight into flux balance during xylitol production from lignocellulosic hydrolysate. Overall elaboration of the strain’s behaviour in multiple feedstock hydrolysates, availability of a toolbox for directed gene deletion/over-expression and transcriptome analysis in lignocellulosic hydrolysates provide early information towards developing a new microbial chassis for bulk chemical production from hemicellulosic carbohydrates.

Professor Gill Stephens Professor of Bioprocess Engineering, University of Nottingham [email protected] https://www.nottingham.ac.uk/Engineering/Departments/Chemenv/People/gill.stephens Research Interests: Biocatalytic methods to produce chemicals, primarily from renewable feedstocks. Current applications-focussed projects include: Metabolic engineering to produce bulk chemicals and monomers from renewable feedstocks; Overcoming product inhibition in microbial processes; Continuous bioprocesses for chemicals manufacturing; Bioproduction of methylmethacrylate.

Bioproess Design: Bio-based methacrylates Methylmethacrylate (MMA) is the monomer used to manufacture Perspex and a wide range of other materials, and is manufactured from petrochemical feedstocks on a scale of 2 million tonnes per annum. As a more sustainable alternative, we investigated bio-based routes to produce MAA from renewable feedstocks. MMA can be produced readily from MAA, using an established chemical esterification process. Therefore, we focussed on bioproduction of MAA. A combined bio-/chemo-catalytic route was developed to produce MAA by engineering an Escherichia coli strain to produce citramalic acid, through overexpression of citramalate synthase, and deletion of the ldhA and pflB genes. The citramalate could be converted to methacrylic acid (MAA) in hot, pressurized water. We also constructed an E. coli strain that could produce MAA directly. Methacrylyl-CoA (MAA-CoA) occurs naturally, as an intermediate of the valine I degradation pathway, so we identified a thioesterase that could catalyse the hydrolysis of MAA-CoA to MAA, and co-expressed it with an acyl-CoA oxidase, branched chain keto-acid dehydrogenase and enzymes from the valine biosynthesis pathway. Therefore, the new bio-based routes to MAA form a promising basis for the development of sustainable MMA manufacturing.

Mr Stuart West Managing Director, Biocatalysts Ltd [email protected] www.biocatalysts.com Research Interests: Enzyme discovery, metagenomics, microbial hosts for enzyme production, development tools for enzyme production scale up.

Rapid enzyme discovery and scale up The world of enzyme discovery and development has totally changed with easy access now to literally millions of enzymes. Samples of novel enzymes can now be produced in a few weeks instead of a few years and at a cost not that different to buying an enzyme from a research biochemical catalogue. Production scale-up can also be done in a much shorter timescale and new laboratory tools are becoming available to make this even faster. This presentation will cover these new developments and highlight where the critical scale up points exist now and the current research challenges.

Dr Georg Lentzen Director R&D, Isobionics [email protected] www.isobionics.com Research interests: Biotechnological production of specialty chemicals with metabolically engineered strains; bioprocess technology and biotechnological manufacturing; isoprenoids; flavours and fragrances, cosmetic ingredients

Microbial production of Flavour and Fragrance isoprenoids Terpenes are used for a large number of applications, including flavours & fragrances, pharmaceuticals, vitamins, pigments and chemical building blocks. Isobionics uses a fermentation-based process for the manufacturing of flavour and fragrance (C10, C15, C20) terpenes, using Rhodobacter sphaeroides. This highly metabolically versatile microorganism has an intrinsic DXP pathway for the provision of the universal terpene precursors IPP and DMAPP. To increase the flux towards isoprenoid precursors, the strain was complemented with genes from the MVA operon from Paracoccus zeaxanthinifaciens. For the production of a specific terpene target compound, a high-performing plant terpene synthase is integrated into the strain. Expression and catalytic activity of the terpene synthase as well as the flux towards terpene precursors are optimized to achieve high-level production of the desired terpene product. In parallel to the strain engineering, the fermentation bioprocess is optimized and adapted to improved strains. A tailored downstream processing yields the F&F terpene product in high yield and constant quality. By plugging in specific terpene synthases into the strain platform, a wide range of different terpenes can be made available using the Rhodobacter platform. This enables the fast growth of a portfolio of commercial products which currently comprises the Citrus ingredients Valencene, Nootkatone, beta-Elemene and beta-Bisabolene.

Dr Alejandro Fernandez-Martell PDRA, University of Sheffield [email protected] Research Interests: My research interest centres on cancer metabolism and directed evolution with particular emphasis on studying the metabolic reprograming –including mitochondrial function, morphology, and capacity– associated with cell differentiation, cell expansion and adaptation, and on the establishment of more accurate and comprehensive cellular models to develop better therapies for neurodegenerative disorders.

Design of mammalian cell factories by direct evolution Mammalian cell have become the predominant factories for the production of therapeutic proteins. However, the complexity and variety of new therapeutic biologics is expanding and continuously imposing major biological constraint to CHO cells that must be controlled to improve the quality of these biopharmaceuticals. Directed evolution has been appreciated as a powerful and fast strategy to come up with a new mammalian cells phenotypes with desirable traits. Here, we directed evolution regimen to engineer biomass intensive CHO cells with increased culture performance and productive state, also, we explore who this approach allow us to improve industrial phenotypes.

Dr Naglis Malys Senior Research Fellow, University of Nottingham [email protected] https://www.nottingham.ac.uk/life-sciences/people/naglis.malys Research Interests: Systems and synthetic biology, as well as molecular mechanisms controlling gene expression. Metabolic engineering and application of synthetic biology tools to develop microbial strains of Cupriavidus necator for production of high value chemicals such as mannitol, 2,3-, 1,3- and 1,4-butanediols, and isoprene.

Engineering bacteria for isoprene biosynthesis Isoprene, a C5 hydrocarbon, is produced by a variety of organisms. As an industrial chemical, it is being used for production of elastomers, adhesives, copolymers and, most notably, as a precursor for synthetic rubbers. The isoprene is produced naturally by plants in large quantities, whereas its current industrial production is based on thermal cracking of oil products. As utilisation of plants for isoprene production is unfeasible and petrochemical sources become more expensive and limited, a sustainable alternative, such as the microbial fermentation-based process, is required for isoprene production. The research in the Synthetic Biology Research Centre (SBRC) focusses on the development of synthetic biology tools and metabolic engineering for production of platform chemicals. Gram-negative betaproteobacterium Cupriavidus necator H16, which directs a high proportion of carbon flux towards production of poly-3-hydroxybutyrate (PHB) and can grow autotrophically by using carbon dioxide and hydrogen as sole carbon and energy sources, is being used as a metabolic chassis. The ability to fix carbon dioxide and feasibility to redirect the carbon flux from PHB to value-added chemicals opens avenues for use of this microorganism as sustainable biotechnology platform. One of research streams at the SBRC is focussed on developing C. necator H16 strains for sustainable bioproduction of isoprene using carbon dioxide as sole carbon source.

Dr Tithira Wimalasena Senior Scientist, Calysta UK [email protected] www.calysta.com Research Interests: Process development, Fermentation, Synthetic biology, Methylococcus, Microbiology.

Introducing Calysta UK Calysta develops and produces high quality protein for commercial aquaculture and livestock feed. Calysta has established its first Market Introduction Facility (D-loop Pilot scale fermentation facilities) in UK for FeedKind® protein, a new sustainable fish feed ingredient to reduce the aquaculture industry’s use of fishmeal. The facility opened in September 2016 and is located at Wilton centre at Teesside, in northeast England. Calysta has also announced a partnership with Cargill for production of FeedKind Aqua protein in North America and marketing worldwide.

Dr John Heap Dr John Heap Lecturer in Synthetic Biology, Imperial College London [email protected] heaplab.org Research Interests: Synthetic biology, metabolic engineering, Clostridium, cyanobacteria.

Technologies for rapid cell factory engineering Strain engineering is required to realise the huge potential of microbial metabolism for industrial biotechnology. Advances in synthetic biology have benefitted the challenging field of metabolic engineering, but most of these benefits have been limited to model organisms like E. coli and S. cerevisiae. Non-model organisms including Clostridium spp. and cyanobacteria have many advantages and great potential. In many such organisms, synthetic biology resources including methodologies, design principles and collections of characterised ‘parts’ (such as promoters) are lacking. We are developing such synthetic biology technologies and applying them to metabolic engineering of ‘exotic’ organisms to produce additional products, decrease the formation of unwanted products, and improve other industrially-relevant properties. Along the way, this work sometimes reveals or hints at interesting underlying biological mechanisms and guides the development of design principles. Recent progress will be described.

Professor Gudmundur Oli Hreggvidsson Group leader in Biotech at Matis & Professor in Microbiology and Biotechnology University of Iceland [email protected] www.matis.is Research Interests: Microbiology & bioprospecting: Genomic and metagenomic bioprospecting of geothermal and marine biotopes. Metabolic engineering: Thermophilic biorefinery organisms for production of platform chemicals, and energy carriers from macroalgal and lignocellulose carbohydrates. Enzyme development: Carbohydrate active enzymes and “molecular enzymes” for genetic engineering; Molecular microbial ecology: physiological, genomic, metagenomic and pangenomic approaches.

Metabolic engineering of Thermophiles and genetic tools Conventional mesophilic organisms such as S. cerevisae and E. coli have limited possibilities in utilizing second generation biomass and have confined process conditions to a narrow range of environmental variables. Progress has also been hampered by the lack of suitable robust species that are genetically amenable as well as having inherent substrate utilization capabilities and product range potential. We are currently working with versatile thermophiles Rhodothermus marinus (an aerobe) and Thermoanaerobacterium AK17 (an anaerobe), as potential biorefinery species capable of utilizing next generation biomass. The work reported describes our approaches and initial results in developing genetic tools and overcoming metabolic bottlenecks for growth, high titre and efficient production of target added value products.

Dr Femi Olorunniji Senior Lecturer in Biotechnology, Liverpool John Moores University [email protected] https://www.ljmu.ac.uk/about-us/staff-profiles/faculty-of-science/pharmacy-and-biomolecular-sciences/femi-olorunniji Research Interests: Characterisation of site-specific recombinases and their applications in the development of synthetic biology tools for DNA rearrangements, logic gates, and genetic switches. Developing serine integrases as tools for rapid assembly and modification of metabolic pathways and for rapid and specific genome engineering in industrial microorganisms. We actively engage in collaborations to explore industrial applications of our findings.

Serine Integrase Recombinational Assembly (SIRA) for rapid multipart DNA assembly and rearrangement The process of assembling multiple DNA fragments into functional gene arrays in plasmids or other extrachromosomal vectors is an important step in engineering novel functions in living systems. This is often a rate-limiting step in metabolic pathway engineering of industrial microorganisms. Recently, several techniques have been developed to speed up this process. Serine Integrase Recombinational Assembly (SIRA) is a versatile and efficient method that facilitates rapid and orthogonal assembly and modification of metabolic pathways. This approach uses phage integrases, enzymes that carry out recombination reactions between short, specific DNA sequences known as att sites. Products of SIRA can be modified by targeted replacement of parts catalysed by the integrase and its recombination directionality factor (RDF). Fusion recombinases in which the integrase is covalently attached to its RDF designed to enhance the efficiency and specificity of reverse recombination reactions have expanded the application of serine integrases in synthetic biology. This talk will describe a model system that illustrates the use of SIRA in biotechnology and synthetic biology.

Dr Naseem Guar Team Leader, International Centre for Genetic Engineering and Biotechnology, India [email protected] , [email protected] http://www.icgeb.org/naseem-gaur.html Research interests: My research interest is to develop novel yeast strains for production of bio-based chemicals and fuel molecules by using low cost carbon sources; Strains development for C5/C6 co-fermentation, Single cell oil production, Expression and secretion of cellulases, Metagenomics for biofuels, Membrane lipids and transporters.

Developing yeast strains for biofuel production Microbial cell factories are excellent alternative for manufacturing bio-based chemicals including biofuel molecules. The advantage of microbial cell factories is that they can use low value materials (agricultural waste) as carbon source for growth and synthesis of desired products. Thus, microbial cell factories have the potential to make major contributions to economy by extracting value from waste materials. To this end, we exploit synthetic biology and metabolic engineering approaches to develop genetically modified strains. C5/C6 co-utilizing yeast strains and FAFE producing strains were recently developed. The laboratory is producing mono component recombinant cellulases and S. cerevisiae strains secreting cellulases were evaluated in simultaneous saccharification and fermentation processes. We use metagenomics approaches to identify novel biocatalysts from the gut microbiomes of herbivorous animals. To understand the role of membrane lipids and transporters in cell factories, we generated a library of ABC transporters and sphingolipid pathway gene mutants in Candida glabrata to evaluate their role in transport across the membrane.

Dr Jon McKechnie Assistant Professor, University of Nottingham [email protected] Research Interests: Life cycle assessment, Techno-economic analysis, Application to biomass-based products and processes

Techno-economic and life cycle environmental aspects of bio-based products Techno-economic analysis (TEA) and life cycle assessment (LCA) tools play important roles in evaluating the potential viability of emerging technologies, extending fundamental science and engineering research to help to understand how systems could perform if deployed. In this talk, I will discuss how TEA and LCA can be applied throughout the technology development cycle. “Prospective” evaluations of innovations being developed in the laboratory are important for identifying technology development targets to achieve viability from technical, financial, or environmental perspectives. At demonstration scales, these tools can help industry to identify potential markets and to better understand implications of system variables on the overall financial and environmental viability of technology deployment scenarios. Finally, LCA can be employed by policy makers to help to ensure that net benefits are achieved by commercialised bio-based products.

Professor Susan Molyneux-Hodgson Professor of Sociology, University of Exeter [email protected] Research Interests: Sociology of science and technology; interdisciplinarity; academic-industry interactions; science & society.

Mobilising ideas of responsible innovation New modes of production and novel processing techniques are viewed as necessary to shift consumer societies to a more sustainable footing. Synthetic biology, as one approach to facilitate this shift, has generated much interest amongst government, industry and various special interest groups. The social and ethical challenges raised by synbio and other such approaches similarly elicit interest from diverse groups in wider society. The notion of 'responsible research and innovation' (RRI) has gained traction at a range of levels within the space of ‘science & society’. What is currently lacking, however, is an understanding of what RRI means in practice. Consequently, there is a limited sense of the efficacy of RI as a policy/tool/resource e.g. we do not understand the extent to which such a notion meets the needs of industry or academia and we have limited understanding of RI’s validity in social terms. An overview of RRI ideas and how these may be taken up, negotiated and re-shaped will be presented.

Lucy Montgomery Bioeconomy Consultant, NNFCC [email protected] http://www.nnfcc.co.uk/ Research Interests: Bioeconomy consultant researching technical, regulatory and market barriers that prevent products obtained from biotechnology or biomass from reaching commercialisation. Background in bioprocess engineering and microbial biotechnology.

Business services for SMEs in the bioeconomy: BioBase4SME and SuperBIO Small companies developing technologies in the bioeconomy often struggle to obtain funding and bring products or processes to the market. SuperBIO and BioBase4SME are two EU projects that address this problem by offering funded services to SMEs active in the bioeconomy. The services include proof-of-concept bioprocess scale-up at BioBase Europe Pilot Plant in Ghent (BE), scale-up and screening at Toulouse White Biotechnology (FR), and testing the final application of biobased dyes, bioplastics and fibres at various centres in North Brabant (NL). There is also a range of non-technical services including IP support, market research, techno-economic assessment, LCA, and evaluation of relevant regulations and green certificates. Support for business plans and help accessing investors is also given. These services aim to provide SMEs with supporting technical, regulatory and financial information that helps them secure the confidence of investors, collaborators and customers.

POSTERS

Professor Colin Harwood Professor of Molecular Microbiology, Newcastle University [email protected] http://www.ncl.ac.uk/cbcb/staff/profile/colinharwood.html Research Interests: Development of tools for refactoring of members of the genus Bacillus using synthetic biology approaches and a range of molecular biological tools. Fundamental and applied aspects of protein secretion in members of the genus Bacillus.

A functional model of the Gram-positive bacterial Signal Recognition Particle and potential biotechnological applications There is a great deal of uncertainty about the intracellular events associated with bacteria protein secretion. In E. coli the three key intracellular players are the Signal Recognition Particle (SRP – a ribonucleoprotein complex of 4.5S RNA and Ffh), SecA and SecB. It is now generally accepted that the SRP is required for the secretion of integral membrane proteins, while the SecA pathway (with or without SecB) is required for proteins that are targeted beyond the cytoplasmic membrane. SRP and SecA/B both function to maintain their preprotein substrates in the secretion competent (essentially unfolded) state required for translocation through the narrow pore of the Sec translocase. In Gram-positive species SecB is absent and the RNA component of its SRP (scRNA, 271 nt) is significantly longer than of its E. coli counterpart (114 nt). Consequently, unlike E. coli, It forms an Alu domain, responsible in higher organisms for translational arrest and subsequent co-translational translocation. The presence of Alu domains in Eukaryal, Archael and Bacillus SRP RNA, but not E. coli, raises fundamental questions about how the SRP functions in bacteria. In particular, do Gram-positive and Gram-negative bacteria translocate their cargo co-translationally and, if so, do they use similar mechanisms? To address this question we have undertaken a detailed analysis of the composition of the Bacillus SRP, using a combination of bacterial-2 hybrid (BACTH), electromobility shift (EMSA) and microscale thermophoresis (MST) assays. We have also shown that a recently identified protein component of the SRP interacts with a key protein involved in ribosomal function, and provides an attractive model for triggering translational arrest. We will discuss the potential biotechnological applications of this model.

Dr Gajendar Komati Reddy PDRA, The University of Manchester [email protected] Research Interests: My research interests are metabolic engineering of microorganisms and the use of synthetic biology tools to engineer a chassis for improving production of valuable chemicals.

Chassis engineering for higher production of monoterpenoids in E. coli Monoterpenes are highly lipophilic and accumulation in biological membrane has several consequences as they interfere with the membrane’s ability to act as a barrier, and interrupt key processes such as transport and energy transduction. An effort to increase production levels and reduce the toxic effects associated with increased production; a chassis organism will require a mechanism to tolerate high levels of monoterpenes. Modifying the membrane and introducing transporters to eliminate the toxic products is a feasible approach for improving monoterpene productivities. In nature, some bacteria produce pentacyclic triterpenoid lipids (i.e. hopanoids) which have sterol-like functions in bacteria thus safeguarding in numerous stress conditions. Hopanoid pathway enzymes, squalene synthase (sqs) from Methylococcus capsulatus and squalene – hopene cyclase (hopE) from Streptomyces peucetius, were cloned and expressed in E. coli. Both proteins were soluble and active in E. coli. We were able to detect squalene, 17-Isodammara-12, 24-diene, 17-Isodammara-20 (21), 24-diene (hopene), in total lipid extracts when the two enzymes were expressed. Currently the localisation of the produced hopene is under investigation. The wild type and hopanoid engineered E. coli will be compared for monoterpenoid production by introducing the monoterpenoid biosynthesis pathway.

Dr Alejandro Fernandez-Martell PDRA, University of Sheffield [email protected] Research Interests: My research interest centres on cancer metabolism and directed evolution with particular emphasis on studying the metabolic reprograming –including mitochondrial function, morphology, and capacity– associated with cell differentiation, cell expansion and adaptation, and on the establishment of more accurate and comprehensive cellular models to develop better therapies for neurodegenerative disorders.

Metabolic Evolution of CHO Cells In this study, we investigate how CHO cell lines create and maintain cellular biosynthetic capacity during fed-batch culture to achieve the optimal combination of rapid exponential proliferation and extended maintenance of high cell biomass concentration. We perform a comparative meta-analysis of mitochondrial and glycolytic functions of 22 discrete parental CHO cell lineages varying in fed-batch culture performance to test the hypotheses that (i) “biomass-intensive” CHO cells exhibit conserved differences in metabolic programming and (ii) it is possible to isolate parental CHO cell lines with a biomass-intensive phenotype to support fed-batch bioproduction processes. We show that for most parental CHO cell lines, rapid proliferation and high late-stage culture performance are mutually exclusive objectives. However, quantitative dissection of mitochondrial and glycolytic functions revealed that a small proportion of clones utilize a conserved metabolic program that significantly enhances cellular glycolytic and mitochondrial oxidative capacity at the onset of late-stage culture. We reveal the central importance of dynamic metabolic re-programming to activate oxidative mitochondrial function as a necessary mechanism to support CHO cell biosynthetic performance during culture.

Dr Farnaz Yusuf National Postdoctoral Fellow, International Centre for Genetic Engineering and Biotechnology, New Delhi-India [email protected] https://www.icgeb.org/yeast-biofuel-group.html Research Interests: Metabolic engineering and synthetic biology, focused on understanding and improving the xylose metabolic pathway in S. cerevisiae for efficient and rapid utilization of xylose for bioethanol production. My research aims to construct a yeast strain for converting lignocellulosic biomass into sustainable fuels and developing this yeast into a more robust, inhibitor-tolerant cell factory.

Engineering natural isolates of Saccharomyces cerevisiae for C5/C6 fermentation for biofuel production The depleting fossil fuel reserves, rising energy demands and the global climate change due to emission of greenhouse gases has switched the focus to the production of biofuels. Biofuels offer a promising cleanest renewable alternative source of energy to meet the energy consumption across the globe. Sustainable biofuel production from renewable biomass requires the efficient and complete use of all abundant sugars in the lignocellulosic biomass. Xylose is the main pentose and second most abundant sugar after glucose in lignocellulosic material. Saccharomyces cerevisiae lacks xylose metabolizing genes therefore, efforts are being made to engineer S. cerevisiae strains for efficient xylose fermentation to make the process economically feasible. The present study aims to construct a xylose fermenting yeast strains with engineered oxido-reductative pathway for xylose metabolism. The study also shows the effect of different promoters on xylose fermentation. The engineered strain is able to grow on xylose as sole carbon source with the maximum ethanol yield of 0.26g/g xylose and productivity of 0.07g/l/h at 96 hours. The further improvement in strain development involves over expression of pentose phosphate pathway and protein engineering of xylose reductase/xylitol dehydrogenase to change their cofactor specificity which is considered as one of the bottlenecks in efficient xylose fermentation.

Mr Alexander Marcus William Van Hagen PhD Student, University of Nottingham [email protected] Research Interests: Renewable energy and biosynthetic chemicals.

Engineering Enzymes to Improve Ethylene Production in Cupriavidus necator Currently we are dependent on non-renewable fossil fuels for energy and to produce chemicals and plastics, which has resulted in the release of large amounts of greenhouse gases, significantly increasing global temperatures. We urgently need to find sustainable alternatives. Ethylene is an industrially significant platform chemical used to produce a wide variety of plastics and chemicals. It is responsible for 1.5% of the USA’s carbon footprint. There are a diverse number of Ethylene-forming enzymes (EFE), predominantly found in pathovars of Pseudomonas syringae. EFE from P. syringae pv. phaseolicola PK2 (EFEP) produces ethylene in a variety of heterologous hosts.Enzyme engineering techniques are required to improve the performance of the enzyme for ethylene production. We have also demonstrated that co-expression of protein chaperones with EFEP improves ethylene production in Cupriavidus necator. The effect on EFEP solubility will also be assessed using these co-expressed chaperones as the solubility of EFEP in heterologous host is a critical factor.

Mr Sean Craig PhD Student, University of Nottingham [email protected] Research Interests: Metabolism, antimicrobial resistance, biofuels, biosynthesis, electron transport chains, renewable energy technology, bacterial membrane translocation.

Towards Photosynthetic Hydrogen production The adverse effects of climate change can only be avoided by decarbonisation of the transport and energy sectors. Hydrogen holds a lot of promise for an alternative energy carrier, however traditional methods for large scale hydrogen production rely on fossil fuels which is both unsustainable and environmentally unfriendly. The current global market is currently estimated at $117.9 billion, which is expected to grow to $152 billion by 2020 (Nagarajan et al., 2017). Microbes such as cyanobacteria offer a sustainable method of hydrogen production with minimal environmental impact, making them an extremely attractive proposition. Cyanobacteria can possess two functionally distinct [NiFe] hydrogenases: an uptake enzyme, only capable of hydrogen oxidation and bidirectional enzyme capable of reducing protons to evolve hydrogen (Ludwig et al., 2006). Synechocystis sp. PCC 6803 encodes one bidirectional [NiFe] which is a heteropentameric enzyme composed of a hydrogenase module (HoxH & HoxY), forming the catalytic core and the diaphorase module (HoxE, HoxF & HoxU). We have previously demonstrated that the hydrogenase of Synechocystis sp. PCC 6803 is thylakoid associated. There are two distinct hydrogenase populations, one dispersed throughout the thylakoid and the other forming distinct puncta, which correlate with hydrogen evolution (Burroughs et al., 2014). We are currently investigating co-localisation of the hydrogenase complex with potential partners from the electron transport chain.

Miss Sophie Vaud PhD Student, University of Nottingham [email protected] Research Interests: I am interested in novel genetic engineering tools for genome-scale engineering and reshaping of strain metabolic pathways. These involve the renowned CRISPR:Cas9 technique and other associated ribonucleprotein complexes like Cpf1 or C2c2 as well as homologous recombination-based technologies.

Development of Genome Engineering tools for Cupriavidus strains The chemoautotrophic bacterium Cupriavidus necator H16 (formerly known as Ralstonia eutropha H16) can utilise H2 and CO2 from syngas as sole energy and carbon sources to grow under aerobic conditions. It also accumulates naturally in large amounts polyhydroxybutyrate, a polyalkanoate used to manufacture biodegradable plastics. This microorganism is of great industrial interest as engineering its metabolism could lead to the production of other highly valuable compounds directly from industrial gas waste. One of these compounds is ethylene, the simplest molecule of the alkene family widely used in the chemical industry (its worldwide production was estimated over 150 million tonnes in 2016). To reshape efficiently the metabolism of Cupriavidus necator and other related strains of the Cupriavidus genus, it is essential to develop fast, flexible and user-friendly genome editing methods. The implementation of innovative genomic engineering in Cupriavidus necator strains combining CRISPR/Cas9 and scarless homologous recombination will be discussed. In addition, first results of ethylene production in Cupriavidus metallidurans, a heavy metal-resistant Cupriavidus strain, will be shown.

Aritha Dornau PhD Student, University of York [email protected] https://www.linkedin.com/in/aritha-dornau-b6331669/ Research Interests: I am a microbiologist with a primary interest in biomanufacturing and bioprocess engineering using waste feedstocks. My PhD project aims to investigate the potential of organic municipal solid waste (OMSW) as a feedstock for producing renewable fuels and chemicals.

Rubbish to resource: growing Escherichia coli on municipal solid waste Population growth, urbanisation and increasing economic prosperity are escalating the rate of municipal solid waste (MSW) production. Globally, the majority of MSW ends up in landfills or is incinerated, polluting the environment and contributing to climate change. On average 40% of MSW is biodegradable and primarily of plant origin, making it an abundant and renewable source of lignocellulose for biorefinery applications. This BBSRC industrial-CASE project aims to investigate the potential of the organic fraction of MSW (OMSW) as a renewable feedstock for biofuel and biochemical production. We work with an organic fibre derived from autoclave pre-treated MSW, produced by our industrial partner Wilson Bio-Chemical (www.wilsonbio-chemical.co.uk). We have characterised the growth of ethanologenic Escherichia coli LW06 on high-sugar hydrolysate produced from Wilson Fibre® through enzymatic hydrolysis with the industrial enzyme cocktail Cellic Ctec3 (Novozymes). Growth of E. coli LW06 is limited on pure MSW fibre hydrolysate but high cell densities are achieved when hydrolysate is supplemented with ammonium and phosphate. Future work will focus on further exploring the physiology of E. coli LW06 growing on MSW hydrolysate and evaluating fermentation performance alongside a collection of diverse microbial species in order to identify promising strains for industrial biomanufacturing.

Miss Vera Salgado PhD Student, Nottingham [email protected] Research Interests: Biotechnology, Reactor design, Bacterial fermentation and Strain engineering, especially towards production of toxic products.

Biological production of Acetaldehyde using autotrophic feedstocks Acetaldehyde is an aldehyde mainly used in food and chemical industries, mostly as an intermediate in synthesis of other chemicals. It is also used as an additive in dairy products, beverages and others to increase flavour and freshness. This compound usually exists in liquid form, is highly flammable and has a low boiling point (20.5 oC). Due to its high reactivity, acetaldehyde is an excellent platform molecule, replacing petroleum as a chemical feedstock for the synthesis of rubber, plastics and larger molecules. [1] Despite its inherent toxicity, acetaldehyde can be produced from biological sources. Reports in literature show that the engineering of certain microbial species allows the production of this molecule. However, there’s always the danger of a rapid conversion to the less toxic ethanol through native alcohol dehydrogenases affecting the production yields, and the need to use edible feedstocks (e.g. glucose) to achieve high production rates. [2,3] In this work, we report the successful engineering of Cupriavidus strain for the production of acetaldehyde, by exporting a pyruvate decarboxylase enzyme (E.C. 4.1.1.1) to the periplasm thus avoiding the conversion of acetaldehyde to ethanol, using CO2/H2 feedstock. [1] Eckert, M., Fleischmann, G., Jira, R., Bolt, H. M. & Golka, K. Acetaldehyde. (2006). [2] Wecker, M. S. A. & Zall, R. R. Production of acetaldehyde by Zymomonas mobilis. Applied and environmental microbiology 53, 2815-2820 (1987). [3] Zhu, H. Engineering E. coli for co-production of acetaldehyde and hydrogen from glucose. Iowa State University (2011).

Dr Roumiana Todorova Assistant Professor, IBPhBME-BAS, Sofia [email protected] www.bio21.bas.bg Research Interests: Cancer research: Functional molecular interactions in sarcoma. Bioinformatics: protein-protein interactions including oncogenic proteins. Enzymology and thermodynamics: Extremophile proteins and their applications. Natural products from plants: applications in medicine and as food additives.

Potential of Extremophilic enzymes in biotechnology Most industrial extremophile enzymes are recombinant forms produced in bacteria and fungi. The purification of recombinant Extremozymes is simplified by heating of cell extract over 70°C (thermophiles), or by high salt concentration (halophiles). Extremozymes are adapted to work in hard physical-chemical conditions. Efficient enzyme activity of recombinant thermophile enzymes require elevated temperatures (up to 100°C), the halophile enzymes require high salt concentration, while acidophiles and alkaliphiles require extreme low or high pH, respectively. Thermostable cellulases are used in the biotechnology for production of biofuel, biomining, paper, pharmaceuticals, fine chemicals and foods. Microbial lipases (yeast, fungal and bacterial) catalyze the hydrolysis of the ester bonds of triglycerides as well as ester synthesis, inter-esterification, alcoholysis, and acidolysis. Lipases are highly stable in organic solvents, exhibit chemoselectivity, and hydrolyze fatty stains on surfaces, leaving no harmful residue. Thermostable lipases expressed in algae become active at high temperatures and release the fatty acids (previously accumulated during cell growth as part of algal phospholipid membranes) in the high temperature conversion of lipids to fuel. Combinations of extremophile proteins have also further perspectives. Thus, fusions with Sso7d protein from Sulfolobus solfataricus (7 kDa, Tm 98°C) stabilizes the DNA and Polymerase complex during PCR extension in amplifying challenging samples (PCR inhibitors, secondary structure, GC-rich regions, longer amplicons) for molecular diagnostics. New extremophilic expression systems have to be developed to achieve high expression of soluble, stable and functional Extremozymes. Microbial diversity and genomics are used to discover new microbial extremophile enzymes with improved catalytic properties for the Biotechnology.

Mr Rajan Patel PhD Student, University of Nottingham [email protected] Research Interests: My main interests are metabolic engineering, biotechnology and molecular biology. I am particularly interested in autotrophic microbes and how they can be used to produce chemical from C1 gases.

Metabolic engineering to improve ethylene production in Cupriavidus necator There is an urgent need for environmentally sustainable fuels, given the diminishing global reserves of crude oil coupled with the deleterious environmental impact of accumulating CO2. We must find green alternatives to satisfy our energy requirements. The production of bulk chemicals via fermentation of CO2 could lead to a 96% reduction in greenhouse gas production. Cupriavidus necator is a Gram-negative, hydrogen oxidising soil bacterium, which is capable of both heterotrophic and autotrophic growth on CO2. It is capable of producing PHA’s (polyhydroxyalkanoate), which can accumulate to 90% of dry cell weight, making it an attractive chassis for the production of fuels and chemicals. It has become evident in recent years that posttranscriptional regulation mediated by sRNA (small non-coding RNA) is critical to many cellular processes, thus making them powerful tools for metabolic engineering and synthetic biology. We aim to exploit this in Cupriavidus necator by engineering artificial sRNAs (Toehold Switches) as a platform for strain improvement for the production of hydrocarbon-based products such as ethylene. We alter metabolism through the process of genome engineering which plays a pivotal role in improving anaplerotic carbon flux and removing carbon sinks which results in enhanced ethylene production. Rational engineering based on genome scale models provides targets for knockouts and overexpression of enzymes in the ethylene pathway to increase yields and prevent bottlenecks at rate limiting reactions.

Dr Dipankar Ghosh Research Associate, Imperial College London [email protected] Research Interests: High value added Biochemicals and Biofuels generations from Biomass following Synthetic Biology & Metabolic Engineering approaches.

Impact of Microbial transporters on advanced biofuels and biochemical generations Demand for renewable bioresources enhances with the growth of the global population. Microbial engineering has accelerated biofuels and biochemicals generations. However, biomolecule systems design is hindered due to microbial naive physiology i.e. low viability or cellular toxicity towards targeted high value added biomolecules (HVABs). To this end, before biosynthetic pathways designed for industrial HVABs productions can be employed, microbial cells must be equipped with genetic factors necessary to maintain cell life under stressful environments for HVABs productions. Efflux pumps could be a way forward to ensure survival of microbial host in such adverse conditions during HVABs biosynthesis. In this current study, we have tried to elucidate the impact of microbial efflux pumps on HVABs generations.

Mr Pierre Reitzer PhD Student, University of Nottingham [email protected] Research Interests: I have an background in biochemical engineering. My main interests are microbiology, synthetic biology, genetic engineering and metabolic engineering. My PhD is focused on the production of ethylene using the plant pathway in Cupriavidus sp.

Bio-ethylene production using plant pathway in Cupriavidus species Industrial chemistry relies heavily on petroleum as a raw material, thus the need for an environmental-friendly alternative is paramount. Ethylene represents a growing market of more than 150 billion USD; however the main production process involves natural gas cracking, which releases large quantities of CO2. This work focuses on utilising the Yang pathway (ACO/ACS) from plants. We have expressed the genes in 3 different strains of Cupriavidus: C. necator, C. bacillensis and C. metallidurans to produce ethylene. We have already demonstrated that ethylene can be produced in both C. necator and C. bacillensis by expressing both the aco and acs genes from Malus domestica and Solanum lycopersicum. Utilising a minimal salts medium containing fructose and glycerol, we were able to generate 70 nmol/ethylene/OD600. Considerably more than previously reported in both E.coli and Synechococcus sp. PCC 7942. We are currently utilising a combination of metabolic engineering, systems biology and directed evolution coupled with a unique attenuation strategy, to generate a robust couple between product synthesis and biomass growth.

Dr Katalin Kovacs Senior Research Fellow, University of Nottingham [email protected] http://sbrc-nottingham.ac.uk/ Research Interests: Synthetic biology and biological (plant and bacterial) engineering; metabolic engineering of eukaryotic and prokaryotic organisms, focusing on the regulation of interacting and competing metabolic pathways and application of synthetic and systems biology tools for the sustainable production of chemicals and fuels.

Metabolic engineering of Cupriavidus necator H16 for production of platform There is an urgent need to develop technologies for the sustainable production of platform chemicals form cheap and renewable sources. The Synthetic Biology Research Centre (SBRC) Nottingham has set out the challenging task of engineering Cupriavidus necator strain H16 as the main microbial factory for the production of C2 (i.e. ethylene), C3 (i.e.3-hydroxypropionic acid) and higher carbon platform chemicals from CO2 as sole carbon source. To do so, we are using an integrative approach, making use of our in-house genome scale model (GSM), our unique gas fermentation facility and expertise in metabolic engineering. Our first target platform chemicals is 3-hydroxypropionic acid or 3-HP, which can be converted to acrylic acid, methyl acrylate, acrylamide, 3-hydroxypropionaldehyde (3-HPA), and into poly(3-HP) and other biodegradable polymers. Biological synthesis of 3-HP proceeds through several metabolic pathways; two of these were considered for engineering in C. necator H16, as they are the most thermodynamically favourable routes. To date, the pathway proceeding via beta-alanine proved to be the most successful. Synthetic pathways were constructed to increase the carbon flux towards beta-alanine synthesis as well as synthetic pathways for the conversion of beta-alanine to 3-HP. Further synthetic pathways for conversion of 3-HP to higher carbon compounds (i.e.C5) are also being constructed and tested. In parallel, production of 3-HP polymer and p(3-HP)-poly(3-hydroxybutyrate) or p(3-HP)-p(PHB) co-polymers are being investigated.

Mr Christian Arenas Research Assistant, University of Nottingham [email protected] http://sbrcottingham.ac.uk/ Research Interests: My main research has been focused in the genetic engineering of microorganisms for the production of commodities. I’m interested in the production of recombinant proteins and also production of platform chemicals from C1 gases.

The genetic basis of 3-hydroxypropanoate metabolism in Cupriavidus necator H16 The first committed step in 3-HP synthesis is the carboxylation of acetyl-CoA to malonyl-CoA, a reaction that is catalysed by the enzyme acetyl-CoA carboxylase (ACC) and tightly controlled at various levels. The second step is the reduction of malonyl-CoA to 3-HP, a conversion catalysed by the bifunctional enzyme malonyl-CoA reductase (MCR) or, in some archaea, by the combination of two monofunctional enzymes which reduce malonyl-CoA first to malonate semialdehyde and then further to 3-HP. However, the generation of efficient and robust production strains remains a major challenge for metabolic engineering. Here, we focus on the well-studied and genetically amenable ‘Knallgas bacterium’ Cupriavidus necator, which was chosen as a C1-chassis for the production of 3-hydroxypropionic acid (3-HP) and other fatty acid derivatives from CO2 and H2. When testing C. necator for its tolerance towards 3-HP, it was noted that it can utilise the compound as the sole source of carbon and energy. Hence, several genes involved in the degradation of 3-HP were identified and inactivated through in-frame deletion, resulting in strain unable to grow on this compound. However, this strain was still able to co metabolise 3-HP alongside other carbon sources such as fructose or gluconate, necessitating further investigation, including the introduction of additional gene deletions. Finally, genes encoding MCRs from different bacteria and archaea were codon-optimised, assembled into functional operons and screened for efficient expression in C. necator.

Mr Matthew Dale PhD Student, University of Edinburgh [email protected] http://rosser.bio.ed.ac.uk/matthew-dale Research Interests: Engineering microbes (yeast in particular) for the production of medicinally and industrially important chemicals. am currently working on the production of plant triterpenoids in the yeast Saccharomyces cerevisiae. Triterpenoids have diverse applications, but their use is currently limited by low yields when extracting from plants, and the difficulty of chemical synthesis due to their complex structures.

Optimising Triterpenoid Production in Saccharomyces cerevisiae Triterpenoids are a diverse group of plant natural products with applications across diverse sectors, including medicine, food, and home and personal care (e.g. as anti-inflammatories, gelling agents and surfactants). Their low natural abundance makes obtaining triterpenoids by extraction from plants difficult, limiting industrial exploitation. However, microbial production potentially offers a cost-effective means of obtaining triterpenoids to the required abundance and purity. This involves heterologously expressing triterpenoid biosynthetic enzymes in a microorganism of choice, allowing potentially any triterpenoid to be produced. This project aims to optimise triterpenoid production in the yeast Saccharomyces cerevisiae. In the first instance, production of β-amyrin and its carboxyl derivative oleanolic acid will be optimised. These triterpenoids and their derivatives have a range of potential applications, including as adjuvants and in the treatment of liver disorders. By expressing various β-amyrin synthases in S. cerevisiae, the homologue that allowed the highest β-amyrin production was identified. The same strategy was successfully applied to P450s of the CYP716A subfamily, which oxidise β-amyrin to oleanolic acid through a multi-step reaction. Varying amounts of the reaction intermediates (erythrodiol and oleanolic aldehyde) and the final product oleanolic acid were shown to accumulate depending on the P450 expressed. Strains expressing these enzymes can serve as starting points for further optimisation through metabolic engineering, and for the production of more oxidised triterpenoids and their glycosylated derivatives (saponins).

Mr Joshua Gascoyne PhD Student, University of Nottingham [email protected] http://sbrcottingham.ac.uk/ Research Interests: Metabolic pathway engineering, Synthetic biology, Combinatorial engineering, Fermentation

Engineering Cupriavidus necator for the production of 1,3-butanediol using synthetic metabolic pathways Butanediols are widely used in the synthesis of polymers, specialty chemicals and important chemical intermediates. 1,3-butanediol is of particular interest due to the optically active form (R)-1,3-butanediol that is used for synthesis of industrial chemicals and as a key intermediate for beta-lactam antibiotic production. Chemical synthesis often produces a racemic mixture of R- and S- forms, whereas the bio-based enzyme-driven production can achieve a high optical purity of (R)-1,3-butanediol. Engineering microorganisms capable of utilising waste greenhouse gases such as CO2 for the production of butanediols provides a promising solution, reducing crude oil consumption and atmospheric CO2 levels. The widely studied facultative lithoautotrophic bacterium Cupriavidus necator is an ideal candidate due to its capability of reaching high cell densities and widely understood mechanism of using CO2 as the sole carbon source with H2 and O2 as energy sources. Through the expression of heterologous enzymes in combination with gene deletions, we engineer C. necator H16 for production of 1,3-butanediol.

Dr Nikolaos Pantidos PDRA, University of Edinburgh [email protected] http://horsfall.bio.ed.ac.uk/page1/page1.html Research Interests: My research interests include the bioremediation of metal contaminated waste from various sources such as land and industrial effluents. I am also involved in making genetically modified bacteria to enhance bioremediation effects. My work is based around improving circular economy and resource efficiency which are topics that are becoming increasingly important in our everyday lives.

The use of M. psychrotolerans as a biofactory for metal nanoparticles Copper pot stills play a crucial role in the creation of the spirit, which once matured becomes Scotch Whisky. The use of copper pot stills in the production of Scotch Whisky is also enshrined in law as part of the Scotch Whisky Regulations (2009). As a result some of the co-products of distillation can contain low levels of copper. Removal of copper from co-products for re-use is an aim of the industry. Current methods of copper removal from such co-products include electrolysis and membrane filtration which are reported to be impractical and costly. Biological copper ion removal from effluents has been shown to be quite effective. We have developed a biological method for removing copper ions from aqueous media and converting them into copper nanoparticles using Morganella psychrotolerans. Elemental analysis has shown they are composed of elemental copper and possess long-term protection against oxidation. This provides the potential for them to be used in numerous applications such as catalysts, antimicrobials and optics. The pathway for copper nanoparticle synthesis however is not very well understood. The aim of this study is to elucidate the pathway of copper nanoparticle synthesis by our bacterium and manipulate the pathway for enhanced metal recovery using synthetic biology tools, leading to an enhanced strain that can be used for the bioremediation of whisky distillery co-products and beyond. Moreover, the potential use of M. psychrotolerans as a biofactory for cold-active enzymes is also being investigated due to its low growth temperature and ease of culturing.

Dr Joyce Bennett Project Manager, University of York [email protected] www.projectdetox.co.uk Research Interests: On Project DETOX we are interested in determining the basis of the toxic effect of the product or feedstock during bio-based production. We are using a multi-omics approach to understand the stresses incurred during industrially relevant fermentations by identifying changes in gene and protein expression, metabolic pathways and effects on the lipid composition.

Project DETOX: detoxifying bio-based production The potential of much bio-based manufacturing is limited by the yield achievable from cell culture. The toxicity of both the product and the feedstock can restrict growth, and hence production, often making a bio-based approach uneconomical. Project DETOX aims to address this limitation by investigating these toxic effects and stress responses thereby enabling engineering of more tolerant host strains for industrial fermentation. Project DETOX will use multi-omics and membrane structure analysis during bio-production in industrially relevant fermentation conditions to reveal the cellular responses to chemical stress in unprecedented detail. This integrated global analysis of the entire cellular system will identify a full portfolio of toxic effects and adaptations, and will be used to inform the development of solutions to improve tolerance. In addition, a programme of work is being undertaken to ensure these advances are in line with responsible research and innovation (RRI). By integrating RRI into our project we aim to ensure our outputs are most likely to meet the needs of society, for now and the future.

Mr Leonardo Talachia Rosa PhD Student, University of Sheffield [email protected] https://www.linkedin.com/in/leonardo-t-rosa/ Research Interests: Rhodopseudomonas palustris, as a model to study the TTT transporter family network. Focused on characterising high affinity transport systems through bioinformatics, biochemistry, structural and physiological assays, with focus on the soluble components of transport, but currently gaining experience with the membrane components as well.

Structural basis for high-affinity adipate binding to AdpC 1 (RPA4515), an orphan periplasmic binding protein from the Tripartite Tricarboxylate Transporter (TTT) family in Rhodopseudomonas palustris

The Tripartite Tricarboxylate Transporter (TTT) family is a poorly characterised group of prokaryotic secondary solute transport systems, which employ a periplasmic substrate binding-protein (SBP) for initial ligand recognition. The substrates of only a small number of TTT systems are known and very few SBP structures have been solved, so the mechanisms of SBP-ligand interactions in this family are not well understood. The SBP RPA4515 (AdpC) from Rhodopseudomonas palustris was found by differential scanning fluorescence and isothermal titration calorimetry to bind aliphatic dicarboxylates of a chain length of six to nine carbons, with KD values in the µM range. The highest affinity was found for the C6-dicarboxylate adipate (1,6-hexanedioate). Crystal structures of AdpC with either adipate or 2-oxoadipate bound revealed a lack of positively charged amino-acids in the binding pocket and showed that water molecules are involved in bridging hydrogen bonds to the substrate, a conserved feature in the TTT SBP family that is distinct from other types of SBP. In AdpC, both of the ligand carboxylate groups and a linear chain conformation are needed for coordination in the binding pocket. RT-PCR showed that adpC expression is upregulated by low environmental adipate concentrations, suggesting adipate is a physiologically relevant substrate but as adpC is not genetically linked to any TTT membrane transport genes, the role of AdpC may be in signalling rather than transport. Our data expands the known ligands for TTT systems and identifies a novel high-affinity binding-protein for adipate, an important industrial chemical intermediate and food additive.

Mr Steven Bourne PhD Student, Aberystwyth University [email protected] Research Interests: Synthetic biology, gene disruption, genetic modification, CRIPSR, molecular biology, yeast, Candida, Candida tropicalis, fermentation, hyphae formation, light microscopy, fluorescence microscopy

Understanding and controlling the phenotypic switch from a yeast-like to hyphal phenotype in Candida tropicalis for use in Industrial Biotechnology ABER Instruments is a world leader in the development and production of biomass probes for use in Industrial Biotechnology (IB). ABER biomass probes are used extensively in breweries to monitor biomass of budding yeast, but having recently branched out into other applications of IB there is a commercial interest in developing an algorithm for monitoring biomass of filamentous fungi. The aim of this PhD project is to do just that. Candida tropicalis is currently being developed as a fermentation platform for use in IB at Aberystwyth University, and has been shown to exhibit two morphological phenotypes when used in fermentation systems; yeast-like and filamentous. In order to develop an algorithm for monitoring biomass, we first need to be able to understand and control the phenotypic switch from yeast-like to filamentous phenotype. In order to achieve this there are three approaches that this project will focus on; 1) adjusting the growth conditions within a fermentation to induce hyphae formation, 2) changing the growth media to induce hyphae formation, 3) identify potential gene candidates which can be disrupted to induce/prevent hyphae formation. As a result of this project so far we have been able to show that hyphae can be induced by; 1) adjusting growth conditions, such as agitation, 2) altering sugar concentrations in standard growth media. 3) A literature review has identified several potential gene candidates which when disrupted can prevent hyphae formation within liquid culture. With a protocol in place for inducing hyphae, research using the biomass probe can begin.

Dr Mary Ann Madsen Post-doctoral researcher, University of Glasgow [email protected] Research Interests: Genetic and environmental manipulation of cyanobacteria towards high value products. Synthetic biology, industrial biotechnology, molecular tools, metabolic engineering, environmental manipulation, ‘omics.

Molecular tools to engineer cyanobacteria for industrial biotechnology Cyanobacteria are a diverse group of photosynthetic prokaryotes with the ability to produce a wide palette of high-value products for pharmaceutical, nutraceutical, cosmetic and other industrial applications. Importantly, the metabolic activities are achieved with minimal input requirements: sunlight, CO2 and water. Cyanobacteria therefore offer new opportunities to generate a sustainable and multifunctional chassis for industrial biotechnology. As a cell factory chassis, cyanobacteria still lack many of the genetic tools available to other organisms such as E. coli or yeast. We therefore developed a standardised molecular toolbox to introduce synthetic expression constructs, control transcription and translation in cyanobacteria. Paired with computational tools, we can now rapidly engineer cyanobacteria for an array of applications. Additionally, we have developed a variety of defined growth conditions to enhance synthesis of natural products. Combined with RNA-sequencing, the environmental responses enable us to unravel biosynthetic pathways and identify target genes for genetic engineering. In summary, we have made important steps towards the control of cyanobacteria, both genetically and environmentally, to study fundamental processes for practical applications.

Dr Robert Bradley Post-doctoral researcher, Imperial College London [email protected] https://www.imperial.ac.uk/people/r.bradley Research Interests: My research interests are within the field of microbial synthetic biology, and include the development of fundamental genetic tools, the creation of new regulatory systems, and harnessing bio-electrochemical processes for biosensing, bioenergy, and electrosynthesis applications.

Synthetic biology for electrosynthetic bioproduction by Rhodopseudomonas palustris This research programme will develop standardised synthetic biology tools for the bacterium Rhodopseudomonas palustris, enabling the scientific community to harness the organism’s versatile metabolism and electroactive properties for biotechnological applications. In addition to promoter, RBS and transcription terminator libraries, we plan to adapt the CRISPR-dCas9 and TetR-family transcriptional repressors for use in Rh. palustris to allow the creation of synthetic regulatory systems that interface with the host genome. Ultimately, we aim to use these new synthetic biology tools to manipulate the unique combination of native capabilities of Rh. palustris and build a versatile bio-catalytic platform for the sustainable production of chemicals, targeting the production of hydrogen and methane gases and a bioplastic precursor as proof-of-principle products. We will also look to add synthetic regulation and additional capacity to the native electron-uptake system of Rh. palustris, enabling our target products to be produced via electrosynthetic pathways.

DELEGATES IN ALPHABETICAL ORDER First Name Last Name Organisation Position Accommodation Networking

Dinner 5th Dec 6th Dec 7th Dec

Abdulrahman Alessa University of Sheffield PhD Student Yes

James Allen UCL PDRA HH Yes

Claudio Angione Teesside University Senior Lecturer HH Yes

Valentine Anyanwu University of Nottingham PhD Student JONAS Yes

Ian Archer IBioIC Technical Director HH Yes

Christian Arenas University of Nottingham Research Assistant JONAS Yes

Joyce Bennett University of York Project Manager HH Yes

Gary Black Northumbria University Professor HH Yes

Adriana Botes VideraBio Director HH HH Yes

Steven Bourne Aberystwyth University PhD student Booked own Yes

Robert Bradley Imperial College London Post-doctoral Research Associate Booked own Yes

Daniel Brown Croda Europe Research Scientist HH Yes

Samantha Bryan University of Nottingham Lecturer JONAS Yes

Micaela Chacon University of Bath Research Associate JONAS Yes

Andrew Collis GSK Technical Lead Synthetic Biochemistry HH Yes

Elizabeth Court University of Sheffield Research Associate Yes

Jillian Couto University of Glasgow PDRA JONAS Yes

Sara Coyotzi University of Sheffield Visiting Researcher Yes

Sean Craig University of Nottingham PhD Student JONAS Yes

Alexander Cudzich-Madry University of Nottingham PhD Student JONAS Yes

Matthew Dale University of Edinburgh PhD Student JONAS Yes

Tim Davies Rubus Scientific Ltd Director JONAS Yes

Mark Dickman University of Sheffield PI No

William Durham University of Sheffield Lecturer of Biological Physics Yes

Graham Eastham Lucite International Senior Research Scientist HH Yes

Jacob Edgerton University of Leeds PhD Student JONAS Yes

Francesco Falcioni Hypha Discovery Head Of Microbiology HH HH Yes

Alejandro Fernandez-

Martell University of Sheffield PDRA Yes

William Finnigan University of Manchester Postdoctoral Research Associate JONAS No

Philippe Gabant SYNGULON CSO HH Yes

Joshua Gascoyne University of Nottingham PhD Student JONAS Yes

Naseem Gaur

International Centre for Genetic Engineering and

Biotechnology Group Leader HH HH HH Yes

Dipankar Ghosh Imperial College London Research Associate JONAS Yes

Jim Gilmour University of Sheffield Senior Lecturer Yes

Miriam Gonzalez

Villanueva University of Sheffield PhD Student Yes

Jeff Green University of Sheffield Professor Yes

Charlotte Green University of Nottingham PDRA JONAS Yes

Darren Greetham University of Huddersfield Researcher JONAS Yes

Sholeem Griffin Swansea University Research Assistant JONAS Yes

Martin Gustavsson

KTH Royal Institute of Technology

Postdoctoral Researcher Yes

Philip Gwyther University of Sheffield Undergraduate No

Ivan Gyulev University of York PhD Student JONAS Yes

Joanna Harley University of Sheffield PhD Student No

Colin Harwood Newcastle University Professor HH HH Yes

John Heap Imperial College London Lecturer HH Yes

Daniela Heeg CHAIN Biotech Technical Product Manager HH Yes

Peter Henderson University of Leeds Professor HH Yes

Ian Higgins Calysta Analytical Scientist Booked Own

Andrew Hitchcock University of Sheffield PDRA Yes

Matthew Hodges Oxford Biotrans Limited Strategy & Business Development HH Yes

James Hough University of Sheffield PGR Yes

Gudmundur Hreggvidsson Matis & University of Iceland Head of Biotech & Professor HH HH Yes

Mahendra Raut University of Sheffield PDRA Yes

Mohamed Issouf Oxford Biotrans Ltd Fermentation Development Manager HH Yes

Stephen Jaffe University of Sheffield PDRA Yes

Adrian Jervis University of Manchester Senior Experimental Officer No

Emily Johnston University of Edinburgh PDRA JONAS Yes

Nitin Kamble University of Sheffield PhD Student Yes

Ifeyinwa Kanu Heriot Watt University Researcher JONAS Yes

Rahul Kapoore University of Sheffield PDRA Yes

Dave Kelly University of Sheffield Professor Yes

Gajendar Komati Reddy University of Manchester Research Associate JONAS Yes

Katalin Kovacs University of Nottingham Senior Research Fellow JONAS Yes

Preben Krabben CPI Principle Scientist HH HH Yes

Kang Lan TEE University of Sheffield Research Associate No

Georg Lentzen Isobionics BV Director R&D HH HH Yes

Jo Longster University of Sheffield PDRA No

Mike Lynch Duke University Assistant Professor HH HH Yes

Mary Ann Madsen University of Glasgow PDRA JONAS Yes

Naglis Malys University of Nottingham Senior Research Fellow JONAS Yes

Laura Martins University of Nottingham PhD Student JONAS Yes

Jon Mckechnie University of Nottingham Assistant Professor HH Yes

Josie Mcquillan University of Sheffield PhD Student Yes

Russel Menchavez University of Nottingham PhD Student JONAS Yes

Samantha Miller University of Aberdeen Senior Lecturer HH HH Yes

Charlotte Milne Reckitt Benckiser LIMS Data Assistant HH Yes

Susan Molyneux-Hodgson University of Exeter Professor Yes

Lucy Montgomery NNFCC Bioeconomy Consultant HH Yes

Hana'a Muheisen University of Nottingham PhD Student JONAS Yes

Ali Mulakhudair University of Sheffield PhD Student Yes

Alison Nwokeoji University of Sheffield Postdoctoral Research Associate Yes

Femi Olorunniji Liverpool John Moores

University Senior Lecturer in Biotechnology Yes

Jags Pandhal University of Sheffield Lecturer Yes

Nikolaos Pantidos University of Edinburgh Postdoctoral Research Associate JONAS Yes

Ricardo Parra Cruz University of Nottingham PhD Student JONAS Yes

Martina Pasini University of Nottingham Research/Fellow JONAS Yes

Raj Patel University of Nottingham BBSRC DTP PhD Student JONAS Yes

Calum Pattrick University of Sheffield PhD student Yes

Trong Khoa Pham University of Sheffield PDRA No

Vincent Postis Leeds Beckett University Reader HH Yes

Pierre Reitzer University of Nottingham PhD JONAS Yes

Leo Rosa University of Sheffield PhD Student Yes

Luca Rossoni University of Nottingham Research Fellow JONAS Yes

Sarah Ryan Fujifilm Diosynth Biotechnologies Senior Research Scientist HH Yes

Vera Salgado University of Nottingham PhD Student JONAS Yes

Stefan Semerdzhiev University of Glasgow PhD Student JONAS Yes

Holly Smith Green Biologics Limited Head of Fermentation HH Yes

Abhishek Somani Aberystwyth University Postdoctoral Scientist JONAS Yes

Vicki Springthorpe University of York Postdoc JONAS Yes

Graham Stafford University of Sheffield Reader Yes

Gill Stephens University of Nottingham Professor HH HH Yes

George Sutherland University of Sheffield PhD Student Yes

Katie Syddall University of Sheffield Post Doctoral Research Associate Yes

Tolu Taiwo University of Nottingham PhD researcher JONAS Yes

Ian Taylor Senior Scientist Dr. Reddy's Laboratories JONAS Yes

Stan Theophilou BlueGene Technologies Ltd Chief Scientist HH Yes

Gavin Thomas University of York Professor HH HH Yes

Roumiana Todorova Bulgarian Academy of Sciences Assistant Professor HH Yes

Alexander Van Hagen University of Nottingham PhD Student JONAS Yes

Jen Vanderhoven University of Sheffield CBMNet Manager HH HH Yes

Sophie Vaud University of Nottingham PhD Student JONAS Yes

Christian Voigt University of Sheffield Lecturer Yes

Christopher Waite Imperial College London Post-doctoral Research Associate Booked own Yes

Stephen Wallace University of Edinburgh Lecturer in Biotechnology JONAS Yes

Joseph Webb University of Sheffield Post doc Yes

Andy Wells University of Nottingham Project Manager JONAS Yes

Stuart West Biocatalysts Ltd Managing Director HH Yes

Roderick Westrop BBSRC Strategy and Policy Manager IBBE HH Yes

Philip Weyrauch Ingenza Senior Biochemist HH HH Yes

Benjamin Willson University of York Post-Doctoral Research Associate JONAS Yes

Tithira Wimalasena Calysta Senior Fermentation Scientist Booked Own

Andrew Yiakoumetti University of Nottingham Research Fellow JONAS Yes

Farnaz Yusuf

International Centre for Genetic Engineering and

Biotechnology Postdoctoral Fellow HH HH HH Yes

Patricio Zapata University of Nottingham PhD Student JONAS Yes