Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 2nd International Conference on Systems and Synthetic Biology London, UK.

Day 2 :

Conference Series Systems and Synthetic Biology 2016 International Conference Keynote Speaker Richard J Naftalin photo
Biography:

Prof. Richard J Naftalin graduated in medicine from Glasgow University. Following completion of medical registration   at Glasgow Royal Infirmary studied at University College London for an M.Sc in Biochemistry and then at the National Institute for Medical Research Mill Hill, for a Ph.D in Biochemistry.  Joined the Physiology dept at Leicester University in 1968 as a lecturer and after seven years moved to King’s College London Dept of Physiology as Senior Lecturer, appointed as Professor in Physiology 1990. 

Abstract:

A computer model designed to simulate integrated glucose-dependent changes in splanchnic blood flow with small intestinal glucose absorption, hormonal and incretin circulation and hepatic and systemic metabolism in health and metabolic diseases e.g., non-alcoholic fatty liver disease, (NAFLD), non-alcoholic steatohepatitis, (NASH) and type 2 diabetes mellitus, (T2DM) demonstrates how when glucagon-like peptide-1, (GLP-1) is synchronously released into the splanchnic blood during intestinal glucose absorption, it stimulates superior mesenteric arterial (SMA) blood flow and by increasing passive intestinal glucose absorption, harmonizes absorption with its distribution and metabolism. GLP-1 also synergizes insulin-dependent net hepatic glucose uptake (NHGU). When GLP-1 secretion is deficient post-prandial SMA blood flow is not increased and as NHGU is also reduced, hyperglycemia follows. Portal venous glucose concentration is also raised, thereby retarding the passive component of intestinal glucose absorption. Two NASH-related mechanical defects; increased pre-hepatic sinusoidal resistance combined with portal hypertension open intrahepatic portosystemic collateral vessels. The model reveals the latent contribution of portosystemic shunting in development of metabolic disease. This diverts splanchnic blood content away from the hepatic sinuses to the systemic circulation, particularly during the glucose absorptive phase of digestion, resulting in inappropriate increases in insulin-dependent systemic glucose metabolism. This hastens onset of hypoglycemia and thence hyperglucagonemia. The model reveals that low rates of GLP-1 secretion, frequently associated with T2DM and NASH, may be also be caused by splanchnic hypoglycemia, rather than to intrinsic loss of incretin secretory capacity. These findings may have therapeutic implications on GLP-1 agonists or glucagon antagonist usage.

  • Synthetic Genomics| Synthetic Gene Network| Gene Synthesis |Biophysics| Bio-Sensors and Bio-Electronics| Next Generation Sequencing| Computational Systems Biology | Pharmaceutical Biology
Location: Kennedy

Chair

Anthony C Forster

Uppsala University, Sweden

Co-Chair

Martin Falk

Czech Academy of Sciences, Czech Republic

Session Introduction

Anthony C Forster

Uppsala University, Sweden

Title: NextGen SynBio: Synthetic genomics and drug discovery

Time : 09:45-10:15

Speaker
Biography:

Anthony C Forster has discovered the hammerhead catalytic RNA structure, invented external guide sequences for ribonuclease P and created unnatural genetic codes de novo, all of which founded biotech companies. He has published in journals including Cell, Nature and Science, edited volumes of Methods and Biotechnology Journal and coauthored "Synthetic Biology: A Lab Manual."

Abstract:

We have developed simplified, purified, bacterial translation systems to facilitate studies of substrate recognition in protein synthesis and enable new applications. Surprises in translation include slower incorporation of proline and other N-alkyl amino acids and that peptide bond formation, not accommodation, is rate-limiting in dipeptide synthesis. One application is directed evolution in vitro of small-molecule, peptidomimetic drug candidates by redesigning the genetic code for the synthesis and display of polymers containing unnatural amino acids. Fast kinetics with unnatural amino acids has identified the causes of several inefficiencies and lead to ways of improving incorporation yields. Another application is cost-effective, scalable, purified, in vitro translation. The latter was achieved by total synthesis and BioBrick assembly of a 58-kbp module encoding 30 translation factor cistrons, breaking new ground in de novo design for synthetic genomics.

Martin Falk

Czech Academy of Sciences, Czech Republic

Title: Towards a complex view on DNA damage and repair: Epigenetic and spatio-temporal aspects

Time : 10:15-10:45

Speaker
Biography:

Martin Falk has completed his PhD from Masaryk University in Brno, CR. He is the Leader of the Department of Cell Biology and Radiobiology at the Institute of Biophysics of the Czech Academy of Sciences. He has participated in more than 30 papers that concern the role of chromatin structure in regulation of cellular processes. His research interests include DNA damage and repair, carcinogenesis, tumor cells radio-sensitization and radiobiology.

Abstract:

Until recently, mainly the genetic and biochemical aspects of processes in the cell nucleus were studied. Nowadays, in the advanced era of 'omics', we have already obtained substantial information on how dozens of proteins interact in the frame of complex cellular signaling pathways and networks. However, additional levels of complexity, like chromatin spatio-temporal organization, have recently emerged as important regulators of fundamental nuclear processes. In this lecture, we will focus on the maintenance of genome integrity, namely the repair of DNA double strand breaks (DSBs). The integrity of the human genome is continuously threatened by intercellular and environmental factors and DSBs represent the most serious DNA lesions; even a single DSB can initiate cell death or cancer when repaired inaccurately. On the other hand, tumor cells are most efficiently killed by DSBs introduced by radiotherapy or chemotherapy. In the last decade, we studied how DSBs, caused by different radiations (high-LET, low-LET) are being induced and repaired. We will discuss the complexity of DSB repair with emphasis on the question of how chromatin structure (nuclear architecture) influences its mechanism and fidelity. Based on the results obtained, we propose a model of the relationship between the higher-order chromatin structure, DSB induction and repair and formation of (carcinogenic) chromosomal translocations.

Nobuo Fukuda

National Institute of Advanced Industrial Science and Technology, Japan

Title: Selection systems for a-type and α-type yeasts using cell-type-specific transcriptional regulation machinery

Time : 11:00-11:30

Speaker
Biography:

Nobuo Fukuda has completed his PhD from Kobe University. He is a Senior Research Scientist of Biomedical Research Institute, AIST. He has published more than 15 papers in reputed journals.

Abstract:

Sake yeasts belong to the budding yeast species Saccharomyces cerevisiae and have high fermentation activity and ethanol production. Although the traditional crossbreeding of sake yeasts is a time-consuming and inefficient process due to the low sporulation rates and spore viability of these strains, considerable effort has been devoted to the development of hybrid strains with superior brewing characteristics. In the current study, we constructed growth selection systems for ‘a’ and α-type cells derived from parental MATa/α yeasts and confirmed that the generated cells possess suitable mating abilities for the production of hybrid yeasts. To achieve it, we designed suitable genetic circuits for expression of the kanMX4 selection marker gene to permit isolation of ‘a’ and α-type cells, respectively, on solid medium. And also, we prevented autopolyploidization of yeast cells derived from the same parent via forced expression of the a1 or α2 gene to increase hybridization efficiency in crossbreeding. Industrially-used strains Kyokai No. 6 and No. 7 were selected as parental sake yeasts and we generated a hybrid strain, designated K76, using the constructed selection systems, which inherited the genetic characteristics of both parents. Notably, because all of the genetic modifications of the yeast cells were introduced using plasmids, these traits can be easily removed. The approach described here has the potential to markedly accelerate the crossbreeding of industrial yeast strains with desirable properties.

Speaker
Biography:

James M Carothers is currently an Assistant Professor at the University of Washington and an Investigator of the Engineering Biology Research Consortium. Previously, he was a Postdoctoral Fellow with pioneering synthetic biologist Jay D. Keasling at the University of California Berkeley. He has earned his PhD at Harvard University with 2009 Nobel Prize-winner Jack W Szostak. He has a BS in Molecular Biophysics and Biochemistry from Yale. His co-authored papers have been cited more than 1300 times and his recent work in synthetic biology has been recognized by the University of Washington Presidential Innovation Award and the Alfred P. Sloan Research Fellowship.

Abstract:

Programmable RNA devices and systems can be engineered for applications in biosensing, information processing and dynamic genetic control. In principle, by combining advanced computational simulations with massively-scaled experimental analysis we investigate the limits of RNA device and system design and engineer synthetic metabolisms to produce medically and industrially-important materials. Here, I will present results using new designable molecular architectures for engineering ultrasensitive RNA aptamer nanosensors as technologies for parallelized metabolic output profiling, a novel class of riboswitch-aptazymes as dynamic metabolite-responsive feedback controllers and programmable guide RNA (gRNA) expression platforms for implementing complex CRISPR-dCas9-based transcriptional networks.

Speaker
Biography:

Mark S Friddin has completed his PhD from the University of Southampton. He has joined the Ces group at Imperial College London as a Postdoctoral Research Associate working on the CAPITALS program in January 2015. His research interests focus on the microfluidic assembly of model membranes, the characterization of ion channels using electrophysiology and the development of smart synthetic microsystems for drug discovery.

Abstract:

Droplet interface bilayer (DIB) networks are becoming increasingly considered as powerful minimal-tissue constructs, however a bottleneck preventing the advancement of this technology is the difficulty of positioning cell-sized droplets into customizable 3D architectures. We address this problem by showing the use of optical tweezers to precisely assemble individual microdroplets ≤20 µm in diameter into complex user-defined 2D and 3D DIB networks. We achieve this by adding sucrose to the aqueous phase to reverse the refractive index contrast of the water in oil microdroplets. This allows us to directly trap the microdroplets in 3D as opposed to exploiting fluid motion generated by the thermocapillary effect or the Marangoni convection effect as reported previously. DIB connectivity between the optically manipulated droplets was confirmed by demonstrating the interdroplet exchange of calcium ions through alpha hemolysin (αHL) nanopores inserted into the membrane. To showcase our ability to method to assemble single and multilayered DIB networks, we constructed both linear and branched symmetric/asymmetric 2D networks together with a 3D droplet tower composed of 11 cell-sized droplets. To our knowledge, the DIB networks assembled using our technique is among the smallest and most complicated reported to date. We envisage that this technology will pave the way for the development of a new generation of minimal-tissues, smart drug delivery systems and bio-electronic circuits assembled from modular droplet components.

Hector Hugo Caicedo

Pharmaceutical Companies of Johnson & Johnson, USA

Title: Real-world microfluidic analytics applications in the pharmaceutical biotech industry

Time : 12:30-13:00

Speaker
Biography:

Hector Hugo Caicedo holds PhD in Bioengineering from the University of Illinois at Chicago. He is the recipient of MIT, Bogazicy University, Antalya University and Universite Pierre and Marie Cuire Pre-doctoral Fellowships as well as Harvard-MIT/HST Post-doctoral Fellowship. He has several publications including several peer-reviewed papers, one book chapter and a provisional patent application. Additionally, he has been awarded more than 20 recognition awards including three National Science Foundation Fellowships in the US and the Mayor’s Civic Merit Medal of Cali given directly by the President of Colombia. Currently, he is a Scientist and Scholar at Janssen R&D, Pharmaceutical Companies of Johnson & Johnson. His interdisciplinary scientific research interests range from material science and bioengineering to cell biology, medicine and pharmaceutical research. At J&J, he is a Subject Matter Expert in Microfluidics Science & Technology and conducts research on drug lead identification, characterization and optimization. Additionally, he is also a Scholar Trainee at the Corporate Sustainability and Innovation program at Harvard University and serves as the Director of Research and Outreach at the US National Biotechnology and Pharmaceutical Association.

Abstract:

Microfluidics has the potential to influence all subject areas of biotechnology from agricultural and environmental to medical and pharmaceutical biotechnology. Even though translational microfluidics science and technology are still at an early stage of development, the pharmaceutical biotech industry may represent the best sector for microfluidics to thrive. With the advent of single-cell analysis, immunotherapy, genetic engineering, micro-scale immunoassays, precision medicine and disease interception; innovative lab-on-a-chip solutions are shaping biotechnology research in ways that are not possible using traditional methods. These solutions enable scientists to shed light on the mechanisms that regulate a myriad of highly intricate biological processes, in both normal and disease states. Here, we highlight some microfluidics-based science and technology that we have developed to enable cutting edge biomedical research, including microfluidics-based real-time tracking of neuronal protein in isolated axons, SAW technology and method development to support pharmacokinetic and pharmacodynamics studies. We also challenge the current paradigm that is focused on searching for a “killer application” as a response for the poor adoption and commercialization of microfluidics technology beyond academic engineering laboratories. Based on our own scientific experience, both in academic and industry laboratories, we provide several reasons that might explain the poor adoption of microfluidics in mainstream biomedical research and the biotech industry. Additionally, we suggest prospects for future directions that could help address some of the present challenges. The aim is enhanced translational microfluidics research that tackles real-world applications in the life sciences and pharmaceutical biotech industry to help develop innovative biotherapeutics for human healthcare.

Krishnan

Imperial College London, UK

Title: Spatial control of biochemical modification pathways and cascades

Time : 13:00-13:30

Speaker
Biography:

Krishnan is a Senior Lecturer in Chemical Engineering and in the Centre for Process Systems Engineering and affiliated with the Institute of Systems and Synthetic Biology at Imperial College. He has completed his undergraduate studies at IIT-Madras, PhD at Princeton University (both in chemical engineering) and was an Associate Research Scientist in Electrical Engineering at the Johns Hopkins University.

Abstract:

Biochemical modification pathways and cascades are the building blocks of signal transduction and are usually studied, analyzed and understood in temporal (lumped) terms. However it is becoming amply clear that in many cellular contexts, reactions occur at different locations and that the spatial aspects of signal transduction and biochemical pathways in general are especially important. This aspect is however poorly understood for multiple reasons. In this talk we focus on the effect of compartmentalization, which is ubiquitous in cellular systems. The effects of compartmentalization are important in understanding concrete pathways and cellular processes which involve compartmentalization, how this has been employed by evolution and also for designing microcompartments in synthetic biology. In this talk, I will discuss the effects of compartmentalization in a range of biochemical modification cascades/pathways: (1) Enzymatic cascades where the output at one stage is an enzyme for the next stage, (2) Pathways where the output at one stage is the substrate for the next stage, (3) Phosphorelays and (4) Open pathways. In each case, a systematic analysis of the effects of compartmentalization is made by comparing the spatially distributed system (modeled by PDEs) with the colocalized system (modeled by ODEs). Through this systematic analysis, we uncover a whole range of effects of compartmentalization of such pathways. We then discuss the relevance and implications of these results for engineering spatial compartmentalization.

  • Young Research Forum
Location: Kennedy

Chair

Richard J Naftalin

King’s College London, UK

Co-Chair

Roberto Mazzoli

University of Torino, Italy

Speaker
Biography:

Pauli Kallio has completed his PhD at the Department of Biochemistry at University of Turku, Finland (2008), studying the secondary metabolic pathways and enzyme-level determinants behind the diversity of bioactive compounds in Streptomyces bacteria. Currently he leads a Synthetic Biology Group in the Plant Molecular Biology Unit at the University of Turku, funded by the Finnish Funding Agency for Innovation (Tekes), with a focus on biofuel research and synthetic biology applications in photosynthetic cyanobacteria. His primary fields of expertise include molecular biology, in vitro enzymology and bacterial metabolic engineering.

Abstract:

Cyanobacteria are a diverse group of photosynthetic prokaryotes which have been extensively studied for their potential as advanced biotechnological hosts for the production of different chemical compounds, namely biofuels and their precursors. The ultimate advantage of cyanobacterial production platforms would be the capacity to generate desired end-products directly from atmospheric CO2 using sunlight as the sole energy without the need for externally supplied sugar as a substrate. Despite the pre-eminent advantages, the use of such production systems is still limited by inefficiency and low production yields, which render the applications commercially non-competitive. To overcome these barriers, our current research is focused on the development and evaluation of a versatile molecular biology toolbox using synthetic biology approaches, in order to effectively harness the photosynthetic capacity to our advantage. Collectively, we aim at developing a palette of standardized validated tools for the assembly and optimization of complex multi-gene over-expression systems in cyanobacteria, which we currently are missing. This will allow us to find, characterize and compare the most suitable genetic elements and expression strategies for our needs and consequently, to exploit our extensive fundamental understanding on the photosynthetic machinery to re-route the metabolic flux towards the target metabolites more efficiently. In our view, systematic synthetic biology approaches combined with the biosynthetic potential of photosynthetic microorganisms may contribute to the development towards sustainable bioeconomy independent of petroleum-based fuels.

Sasa Rezelj

National Institute of Chemistry, Slovenia

Title: Engineering listeriolysin O for specific pore formation in vitro

Time : 14:50-15:10

Speaker
Biography:

Sasa Rezelj has completed her MSc from Biotechnical Faculty of the University of Ljubljana. She is the currently a PhD student in Department for Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Slovenia. Her area of interest is synthetic biology approach to study and engineer listeriolysin O under supervision of Professor Dr. Gregor Andreluh.

Abstract:

Listeriolysin O (LLO) is the pore forming toxin and the most important virulence factor of bacteria Listeria monocytogenes, which causes food-borne disease listeriosis. It belongs to cholesterol-dependent cytolysins (CDC), a family of pore-forming toxins produced primarily in Gram positive bacteria. LLO enables bacteria to escape from host phagolysosome and to spread into the neighboring cells. Moreover, LLO is a unique protein among CDC, since its stability is pH dependent. Pore forming proteins, such as α-hemolysin, have shown their way to biotechnological applications (e.g., DNA sequencing through nanopore). LLO also found its potential use in medicine as a vaccine adjuvant and carrier molecule for anti-tumor therapies and gene delivery. We believe that by revealing the exact mechanism of LLO pore formation, we can engineer LLO to form pores, which can be manipulated and used in biotechnological applications. First, we used giant unilamellar vesicles (GUVs) to study LLO’s mechanism of pore formation. Confocal fluorescence microscopy and flow cytometry were used to analyze time, concentration and environment dependent pore formation of LLO. We discovered the damaging effect of LLO’s pore formation on membranes by analyzing the permeability of GUVs for fluorescent dextrans (FDs) of different sizes and populations of GUVs upon LLO addition. LLO pore forming activity enabled time dependent permeabilization of FDs up to 150 kDa, which is larger than previously assumed pore diameter. LLO was able to damage the vesicles in such extend, that it led to their destruction. Furthermore, we used cell free in vitro synthetic biology approach to construct and produce LLO inside synthetic phospholipid vesicles, which can be specifically controlled by environmental changes.

Reem Swidah

The University of Manchester, UK

Title: Engineering of Saccharomyces cerevisiae toward n-butanol production

Time : 15:10-15:30

Speaker
Biography:

Reem Swidah has completed her PhD from Manchester University in 2016 and she has been working with her supervisors Mark Ashe and Chris Grant on biofuel production from yeast. She has published her first paper in Biotechnology for Biofuels.

Abstract:

Biobutanol represents a second generation biofuel, which can be produced naturally by a number of microorganisms. This alcohol has a number of significant advantages over bioethanol in terms of its physical properties as a fuel, but production systems suffer from various drawbacks. Therefore, we sought to transplant an entire butanol production pathway (the ABE pathway) into a Saccharomyces cerevisiae strain. However, this pathway was incapable of generating reasonable yields of butanol without further metabolic alteration to channel carbon towards the substrate of butanol production, acetyl CoA. For instance, the major alcohol dehydrogenase, ADH1, was deleted and two enzymes involved in acetyl-CoA biosynthesis were overexpressed to give strains capable of producing 300 mg/L butanol. Surprisingly, deletion of the ADH1 gene alone is sufficient to produce 40 mg/L butanol from an endogenous pathway. Previously, this endogenous butanol production pathway was characterized and proposed to derive from the mitochondrial catabolism of threonine via multiple leucine biosynthetic genes and the conversion of 2-ketovalerate to butanol. Therefore, in the studies described here, we aimed to understand the relative contribution of exogenous and the endogenous pathways of butanol synthesis to overall butanol yields. Work will be presented to suggest that both pathways are active in yeast adh1D mutants, but that the endogenous route for butanol synthesis is weaker and does not use the pathway previously proposed via Leucine metabolic enzymes. This work therefore makes use of synthetic biology and metabolic engineering to effectively set the scene for an initiative towards higher yields of butanol in yeast via concerted interventions in both the endogenous and exogenous pathways.

Speaker
Biography:

Chiara Gandini has completed her MSc in Industrial Biotechnology in 2012. She is Coauthor of an international patent. She has been working in Biochemistry Laboratory of Department of Life Sciences and Systems Biology, University of Torino, on microorganism’s metabolic engineering since 2012.

Abstract:

Consolidated Bioprocessing (CBP) is a key feature of the so-called 3rd generation biorefinery consisting of cost-sustainable single-step (direct) fermentation of cellulosic biomass into industrially relevant products such as biofuels (notably, ethanol and or butanol). No natural microorganism isolated so far can produce biofuels directly from cellulose with the efficiency required by industrial processes. Clostridium cellulovorans is an anaerobic bacterium among the most efficient plant biomass biodegraders. It efficiently hydrolyzes cellulose, xylan and pectin, but its main catabolites are organic acids, while little ethanol is biosynthesized. The final aim of the present study is the development of recombinant ethanol and or butanol hyper producing C. cellulovorans strains by metabolic engineering. Achievement of this purpose is currently hampered by major hurdles. Nowadays, no detailed study on C. cellulovorans central metabolic pathways exists. Furthermore, no specific tools for C. cellulovorans chromosomal gene knock out/in are available. The present contribution was focused on the development of a model of the C. cellulovorans central metabolism by integration of metabolomics, transcriptomics and proteomics data. Attention was mainly given to comparison of C. cellulovorans metabolic network during growth on cellulose with respect to cells growing on soluble sugars. As far as the development of optimized gene tools for C. cellulovorans is concerned, different transformation protocols were compared. Furthermore, the ClosTron methodology was used as a basis for developing reliable protocol for gene integration in the C. cellulovorans chromosome. These results will be a key for future C. cellulovorans metabolic pathway engineering aimed at direct conversion of cellulose to biofuels.

Speaker
Biography:

Loredana Tarraran has obtained her MSc in Molecular Biology from Turin University. She is interested in environmental field and working on metabolic engineering of microorganisms for application in biorefinery since 2009. She is author of one national congress communication and Co-Author of other three national and three international congress communications. Furthermore she is Co-Inventor of one international patent.

Abstract:

Lactic acid (LA) is an extensively employed chemical. Notably, one of its main current applications is for synthesizing biocompatible and biodegradable plastic biopolymers, i.e., polylactide (PLA), poly (lactic acid-co-lysine), poly (lactic acid-co-glycolic acid). The latter can be used for clinical application (i.e., tissue engineering) and especially PLA as packaging thus replacing traditional plastics. Lactic acid bacteria are the main natural LA producers. Both “green” property and cost-sustainability of LA-based polymers can be improved if LA is obtained by fermentation of abundant cellulose-based wastes produced by our society, such as municipal solid waste and agricultural by-products. In this light, the development of a microorganism able to both use cellulose as fermentation substrate and produce LA can achieve both environmentally and economically sustainable biopolymer production. The aim of this work was to obtain a recombinant Lactococcus lactis able to depolymerize cellulose by expressing heterologous proteins derived from the cellulolytic complex, i.e., the cellulosome of Clostridium cellulovorans. C. cellulovorans cellulosome consists of enzyme subunits attached to a non-catalytic scaffold protein. Such scaffoldin is anchored to C. cellulovorans cell surface and allows optimal overall enzyme spatial organization, close to both cellulosic substrate and cell surface thus leading to improved cellular intake of cellulose hydrolysis products. In this work four different recombinant scaffoldins were constructed by molecular assembling of different protein domains of C. cellulovorans cellulosome. Engineered scaffoldins were successfully expressed by L. lactis. Their presence in cytosolic, extracellular and cell-wall fractions of L. lactis and their interaction with cellulosomal enzymes were analyzed by multiple approaches.

Speaker
Biography:

Denis Kalemasi has completed his MSc in Industrial Biotechnology at the University of Turin in 2015. He has been working since 2014 on the construction of a recombinant L. lactis for the direct conversion of cellulose into L-lactic acid.

Abstract:

One of the possible applications of synthetic biology is the “creation” of recombinant bacteria able of producing building blocks. The latter have great potential since they can be used for the production of bio-based polymers possibly replacing oil-derived plastics. Lactic acid (LA) is among the most requested compounds by the chemical industry and is already largely employed for the production of its polyesters (i.e., polylactides), which are general purpose biodegradable and biocompatible plastic materials. Most LA produced worldwide is obtained by lact ic acid bacteria (LAB)-based fermentation. However, current processes depend on expensive feedstocks (e.g., glucose) or compete for food crops (e.g., corn). Cellulose, instead, is an inexpensive substrate found in plant material-derived waste. Our aim is to construct a strain of Lactococcus lactis able to produce LA directly from cellulose (that is without prior enzymatic saccharification by commercial cellulases) by metabolic engineering. Only relatively few natural bacteria are able to grow with only cellulose as the substrate. Clostridium cellulovorans is one of them. To achieve our goal, we’ve introduced components of C. cellulovorans cellulose depolymerizing machinery into Lactococcus lactis. In particular, in this work we have shown that, by introducing only two C. cellulovorans glycosyl hydrolases, we were able to develop a recombinant L. lactis able of metabolizing short chains of cellulose (i.e., cellooligosaccharides with degree of polymerization up to 10) into L-LA with a yield close to 100%.

Speaker
Biography:

Dr. Ingy Moustafa Hashad’s work involved the study of “contribution of the p22 phox gene of NAD(P)H oxidase and eNOS gene polymorphisms in the predisposition of early onset acute myocardial infarction in Egyptian population”. She learnt several techniques through her research including, conventional PCR and SNP analysis, ELISA, electrophoresis, and spectrophotometric determinations. She finished successfully her Ph.D. in May 2012 with great appraisal from the supervisors and examiners. Later, she had the opportunity to spend three months summer research in the research lab of Prof. Wolfgang Poller, Charité Centrum für Herz-, Kreislauf- und Gefäßmedizin and another three months in the research lab of Prof. Michael Bader, Max-Delbruck Center for Molecular Medicine, Berlin, Germany which added to her scientific and technical knowledge and skills.

Abstract:

Background: Cardiovascular diseases (CVD) are the universal cause of morbidity and the leading contributor to mortality in both developed and developing countries nowadays. Connexin (Cx) proteins are the building blocks of gap junctions. Among these, Cx37 and Cx40 have been found to be expressed on vascular system and reported to have a cardio protective role. Glutathione peroxidase-1 (GPx-1) enzyme and Manganese Superoxide Dismutase (Mn-SOD), represent a defense mechanism against oxidative stress, thereby contributing to the prevention of atherosclerosis. Besides, high homocysteine levels (Hcy) and Hexanoyl Lysine adduct (HEL) are considered to be an independent risk factor for wide range of diseases such as CVD and their complications including Acute Myocardial Infarction (AMI). AMI is inflammatory pathology, including cytokines such as Fractalkine which plays a central role in inflammation and tissue injury.

 

Subjects & Methods: A total of 205 Egyptian subjects were recruited for the study. They were divided into 105 AMI patients and 100 healthy controls. Genotypes for each participant were determined using a polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) for the Cx37, Cx40, GPX-1 and Mn-SOD genes. Serum levels of sVCAM-1, HEL adduct, Homocysteine and fractalkine were detected quantitatively using ELISA.

 

Results: Allele frequencies for both Cx37 and Cx40 were not significantly different between AMI patients and controls (p=0.93 and 0.24, respectively). The genotype distribution for GPx-1 gene was not significantly different between the AMI patients (CC 56.7%; CT 41.7%; TT 1.7%) and control subjects (CC 53%; CT 45%; TT 2%), (P=0.6008). The prevalence of the V/V genotype of the Val16Ala of Mn-SOD gene polymorphism was significantly more frequent in AMI patients than in control subjects (p=0.0142). In addition, Serum levels of sVCAM-1, HEL adduct, Homocysteine and fractalkine were significantly elevated in AMI patients compared to control subjects (p=0.0273).

 

Conclusion: Contribution of inflammation and oxidative stress markers in addition to Mn-SOD gene polymorphism in the pathogenesis of AMI in Egyptian Population.

Speaker
Biography:

Azizah Nahari is a PhD student in her last year from the University of Edinburgh. She has a Master’s degree from King Abdulaziz University. She is working as a Lecturer at King Abdulaziz University School of Biological Science.

Abstract:

Limitation of crop productivity by phosphate (Pi) is widespread and will probably increase in the future. A better understanding of phosphate use efficiency (PUE) is required for engineering nutrient-efficient. In this study, we designed a hydroponic system for a specific reason which is to avoid complication from differences in Pi uptake. The current experiment examined 213 of the total 527 MAGIC lines under high and low phosphate conditions in hydroponic system, and the results generated considerable variance with regard to various PUE elements and growth features (namely, RFW, SDW, SPC, and flowering time). By using QTL mapping, we have identified QTLs involved in PUE. Having examined the broad 213 MAGIC population, considerable variance in PUE and its constituent features has been recorded. This is indicative of the manifest variance in PUE for the examined lines. It is significant to note that, at the level of the species, lines under the low Pi condition displayed greater PPUE when considered in relation to those under the high condition. Furthermore, the association with regard to PPUE and the features of SDW and Pi content was determined as positive for all lines under the low Pi condition. Notably, these findings provide valuable insight regarding individual lines and the degree to which they can grow effectively in high and low Pi conditions. Additionally, it has identified lines characterised by dissimilar PUE values that could be appropriate for utilisation in breeding schemes with the intention of enhancing the features in novel cultivars.

Speaker
Biography:

Anna K Stavrinides has completed MSc in Plant Genomics and Crop Improvement and she is currently undertaking PhD in the Lab of Prof Sarah O’Connor both at University of East Anglia and the John Innes Centre. She has Bachelors degree in Molecular Biology from the Université Montpellier 2. Her interests are focused around plant abiotic stress resistance and plant/environment interactions.

Abstract:

Monoterpene indole alkaloids are a diverse family of compounds, some of which are of economic importance as drugs for use in the clinic. The medicinal plant Catharanthus roseus (Madagascar periwinkle) produces more than 100 monotepene indole alkaloids and is the source for two anti-cancer alkaloids vinblastine and vincristine. Biosynthesis of these alkloids is long and involves >20 enzymatic steps and multiple tissues and cell compartments, which have complicated the discovery of the biosynthetic enzymes necessary for production of these useful alkaloids. The central intermediate for generation of the various monoterpene indole alkaloid scaffolds is strictosidine, which can be found in many plants, primarily of the Apocynaceae and Gelsemiaceae families. This alkaloid is deglycosylated and the resulting compound is reactive and unstable. Monoterpene indole alkaloids with various backbones can be obtained from this precursor and are found in different species. Recently we discovered the first enzyme which can take this reactive precursor and reduce it to form a heteroyohimbine backbone (Tetrahydroalstonine synthase). This discovery completes the biosynthesis of an alkaloid found in many plant species which produce strictosidine such as Rauvolfia serpentina. I will present our recent work on the elucidation of this dynamic branch of the monoterpene indole alkaloid pathway. Using the information we have gained about the system (gene expression, protein interaction, localization, enzyme specificity) we propose an integrated hypothesis for the evolution of the heteroyohimbine biosynthetic enzymes in the medicinal plant Catharanthus roseus.