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 1 :

Conference Series Systems and Synthetic Biology 2016 International Conference Keynote Speaker Guido Krupp photo
Biography:

Dr. Jean A. Boutin graduated (Thèse d’Etat en Sciences Biologiques) from Nancy University (France) on drug metabolism. He did his postdoctoral training at Johns Hopkins (Baltimore) and at the Karolinska Institutet (Stockholm, Sweden). He was hired as protein chemist in Les Laboratoires SERVIER (LLS) in 1986. During the 30ish last years, Dr. Boutin moved from oncology to peptide research and then molecular & cellular pharmacology. Recently, LLS created a drug discovery platform that Dr. Boutin leads. This structure involves all the aspects of drug discovery, from molecular modeling to ligand/protein biophysical interaction measurements, including protein chemistry, stem cells, structural biology, chemogenetics, HTS, biophysics, Biologics…The main interests of Dr. Boutin are N-myristoyltransferase, melatonin, quinone reductase 2, MCH and autotaxin. In relation with Biologics, our group more particularly explores all the areas related to the possibilities to incorporate exotic amino acids into proteins, especially enzymes.

Abstract:

Synthetic biology is a growing field in which the contribution brought by chemical synthesis of proteins and particularly enzymes is fundamental. Despite this fact, the chemical synthesis of catalytic active proteins remains poorly documented, essentially because it is hard to obtain enough material to use it in biochemical experiments. Chemical synthesis of proteins could permit to have access to the incorporation of unnatural (exotic) amino acids into catalytic active proteins, a feature amenable by recombinant technologies, but that requires delicate manipulation of the bacterial machinery. The developments brought by this approach, include but are not limited to the measure of the influence of unnatural (exotic) amino acids on the 3D structure of enzyme, its activity as well as their recognition of substrate, co-substrate or regulator. The main limitation remains a quantitative problem: How to progress from microgram of proteins produced nowadays with essentially recombinant techniques to tens of milligrams? We chose to present as a model the synthesis and thorough characterization of calstabin, a short protein proline isomerase of 107 amino acids. The protein was synthesized using the native chemical ligation approach. Several tens of milligrams were obtained. Therefore, we were able to refold the polypeptide properly, to characterize its biophysical properties, to measure its catalytic activity and finally to crystallize it in order to obtain its tridimensional structure after X-ray diffraction. Further to it and as a first step of validation of the whole process, we incorporated exotic amino acids in the easiest reachable part of the protein N-terminus. Avenues are now open to obtain further proteins modified with exotic amino acids in a way that is only barely accessible by molecular biology. We hope that these approaches will permit to gain detailed information on the structure-function relationship of proteins as long as they are reachable by complete chemical synthesis (below 300 amino acids).

Keynote Forum

Michael C Jensen

Seattle Children’s Research Institute, USA

Keynote: Enhancing the synthetic IQ of CAR T-cells

Time : 10:45-11:30

Conference Series Systems and Synthetic Biology 2016 International Conference Keynote Speaker Michael C Jensen photo
Biography:

Michael C Jensen was graduated from the University of Pennsylvania; School of Medicine and completed training in Pediatric Hematology and Oncology at the University of Washington, Fred Hutchinson Cancer Research Center. He is the Sinegal Endowed Professor of Pediatric Hematology-Oncology and Bioengineering at the University of Washington, School of Medicine. He serves as the Founding Director of the Ben Towne Center for Childhood Cancer Research at Seattle Children’s Research Institute. He has been a pioneer in the area of cancer immunotherapy focusing on the genetic engineering of immune system T cells. He is an Inventor on a broad portfolio of patents in this area and leads first-in-man clinical trials of chimeric antigen receptor redirected T-cell immunotherapy. He is also a Scientific Founder of Juno Therapeutics, Inc.

Abstract:

Recent conceptual as well as technological advances in the areas of molecular immunology, gene transfer and cell processing have fostered increasingly sophisticated translational applications of adoptive T-cell therapy for oncologic disease employing genetically-modified T-lymphocytes. My laboratory’s work focuses on T-cell genetic modification for re-directing antigen specificity to tumors utilizing recent advances not only in the composition and specificity of receptor antigen recognition domains but also the evolution of multifunctional cytoplasmic signaling domains developed for these chimeric antigen receptors (CARs) that provide dual activation and co-stimulatory signaling. My group is also investigating the context of adoptive transfer with respect to the conditioning of the recipient for enhanced T-cell engraftment and expansion, the grafting of CARs on to central memory T-cells having endogenous TCR specificities for viral epitopes to which the host has robust immunity, and the provision tumor microenvironment survival capabilities. The increasingly broad array of genetic manipulations including not only transgene insertion but targeted gene knock out using engineered targeted nucleases such as TALEN’s and ZFN as well as expression regulatory constructs provides for the creation of synthetic biology of orthogonal immune responses based on gene modified T-cell adoptive transfer. The next decade of advances in this arena will depend on iterative bench-to-bedside back-to-the-bench translational studies capable of sustaining the evolution of these technologies in the context of clinical parameters relevant to the pediatric oncology patient population.

  • Structural Biology | Integrative Biology | Protein Engineering | Industrial Systems and Synthetic Biology | Biotechnology Advances | Metabolomics
Location: Kennedy

Chair

Guido Krupp

AmpTec GmbH, Germany

Co-Chair

Roberto Mazzoli

University of Torino, Italy

Session Introduction

Jean A Boutin

Institut de recherches SERVIER, France

Title: Use of synthetic proteins and future trends: The example of calstabin

Time : 11:45-12:15

Speaker
Biography:

Dr. Jean A. Boutin graduated (Thèse d’Etat en Sciences Biologiques) from Nancy University (France) on drug metabolism. He did his postdoctoral training at Johns Hopkins (Baltimore) and at the Karolinska Institutet (Stockholm, Sweden). He was hired as protein chemist in Les Laboratoires SERVIER (LLS) in 1986. During the 30ish last years, Dr. Boutin moved from oncology to peptide research and then molecular & cellular pharmacology. Recently, LLS created a drug discovery platform that Dr. Boutin leads. This structure involves all the aspects of drug discovery, from molecular modeling to ligand/protein biophysical interaction measurements, including protein chemistry, stem cells, structural biology, chemogenetics, HTS, biophysics, Biologics…The main interests of Dr. Boutin are N-myristoyltransferase, melatonin, quinone reductase 2, MCH and autotaxin. In relation with Biologics, our group more particularly explores all the areas related to the possibilities to incorporate exotic amino acids into proteins, especially enzymes.

Abstract:

Synthetic biology is a growing field in which the contribution brought by chemical synthesis of proteins and particularly enzymes is fundamental. Despite this fact, the chemical synthesis of catalytic active proteins remains poorly documented, essentially because it is hard to obtain enough material to use it in biochemical experiments. Chemical synthesis of proteins could permit to have access to the incorporation of unnatural (exotic) amino acids into catalytic active proteins, a feature amenable by recombinant technologies, but that requires delicate manipulation of the bacterial machinery. The developments brought by this approach, include but are not limited to the measure of the influence of unnatural (exotic) amino acids on the 3D structure of enzyme, its activity as well as their recognition of substrate, co-substrate or regulator. The main limitation remains a quantitative problem: How to progress from microgram of proteins produced nowadays with essentially recombinant techniques to tens of milligrams? We chose to present as a model the synthesis and thorough characterization of calstabin, a short protein proline isomerase of 107 amino acids. The protein was synthesized using the native chemical ligation approach. Several tens of milligrams were obtained. Therefore, we were able to refold the polypeptide properly, to characterize its biophysical properties, to measure its catalytic activity and finally to crystallize it in order to obtain its tridimensional structure after X-ray diffraction. Further to it and as a first step of validation of the whole process, we incorporated exotic amino acids in the easiest reachable part of the protein N-terminus. Avenues are now open to obtain further proteins modified with exotic amino acids in a way that is only barely accessible by molecular biology. We hope that these approaches will permit to gain detailed information on the structure-function relationship of proteins as long as they are reachable by complete chemical synthesis (below 300 amino acids).

Speaker
Biography:

Roberto Mazzoli has completed his PhD in 2003 and he has been working as an Assistant Professor in Biochemistry at the University of Turin, Italy since 2011. He has been studying the metabolism of microorganisms aimed at industrial processes. His main research activity has concerned metabolic engineering of microorganisms for cellulosic consolidated biorefinery. He is the PI of the group of Proteomics and Metabolic Engineering of Prokaryotes at DBIOS, University of Turin. He has published 21 papers in peer-reviewed international journals, 1 book chapter and 2 international patents and he is serving as Editorial Board Member or Reviewer for several reputed journals.

Abstract:

Cellulosic biomass is the most interesting substrate for biorefinery, owing to its abundance and low cost. However, cellulose is highly recalcitrant to biodegradation. Currently available technologies for fermenting cellulose to industrially relevant compounds (e.g., fuels, plastics) consist of multiple-step processes which are not cost-competitive with traditional petrochemical refinery. Engineering recombinant microbial strains able to catalyze single-step fermentation (i.e., consolidated bioprocessing, CBP) of biomass into high-value products is a key goal so as to develop cost-competitive cellulosic biomass biorefinery. For at least 20 years, intensive research efforts have been dedicated to develop recombinant strains suitable for cellulose-CBP following either the “native cellulolytic strategy”, aimed at engineering natural cellulolytic microorganisms so as to improve their product formation features or the “recombinant cellulolytic strategy” aimed at conferring cellulolytic ability to strains which naturally produce industrially-relevant products with high efficiency. Although these researches have attained impressive results, current forefront achievements yet resemble more to proofs-of-concept than to new biotechnological deliverables ready for application in industrial fermentations. The present contribution aims at providing an overview on most significant successful examples of these strategies together with major issues which still need to be addressed and possible solutions.

Speaker
Biography:

Taizo Uda has obtained his PhD from Kyushu University and Postdoctoral studies from Wisconsin University-Milwaukee. He has studied on heterogeneous catalysis and antibody engineering. He was the Director of Diagnostic Group of a Research Institute in a Chemical Company. He is a Visiting Professor at Oita University and Kyushu University. He has published more than 50 papers in reputed journals.

Abstract:

Issue on the structural diversity (heterogeneity) of the molecule has been focused along with the development of recombinant antibody drugs. The structural diversity provides some (or many) isoforms of an antibody caused by different charges, different molecular sizes and/or modifications of amino acid residues. For practical use, the antibody and/or the subunits must have a defined structure. A whole antibody is consisted of the light and heavy chain. Once they are separated, the structure of the light or heavy chain becomes very flexible, which also causes the structural diversity and then gives some kinds of isoforms of different pI. We prepared several human antibody light chains possessing a C-terminal histidine-tag, which was expressed in E. coli. After Ni-NTA chromatography, the purified light chain was subjected to the cation-exchange chromatography, where several peaks consisted of the monomers and/or dimers were observed at the different retention time. This suggests that the different forms in both molecular sizes and the electrical charges co-exist in the solution, while only a light chain is present. This was the similar results regarding molecular heterogeneity as those observed in recombinant antibody drugs as reported. Several metal ions were examined to investigate the effect on the structural heterogeneity (diversity). Note that copper ion exhibited huge effect for solving the heterogeneity (diversity) issue. In the presentation, the role of the constant domain will be also introduced in detail.

Paul Lomax

TTP Labtech, UK

Title: Automation at the heart of synthetic biology

Time : 14:15-14:45

Speaker
Biography:

Paul Lomax is Product Manager for TTP Labtech, sample management products. With over 15 years of experience in the automation of life science applications, he is responsible for TTP Labtech’s range of novel automated -20°C and -80°C storage systems.

Abstract:

With momentum building in the establishment of Synthetic biology foundries, it is evident that automation plays a key role throughout the Synthetic Biology workflow. Safe, secure and robust inventory storage and management is critical. To maximize efficient operation, rapid access to stock constructs or “biobricks” is key and TTP Labtech offer solutions for automated storage at -20°C and -80°C with true cherry picking ensuring that only the required samples are retrieved. 2D barcoded labware allows samples to be tracked and stored and retrieved with the simplicity and speed of a vending machine. Novel pneumatic technology to move tubes minimizes the need for robotics in the cold zone, providing long term reliability of operation. Learn more about these how TTP Labtech’s solutions could automate the heart of your Synthetic Biology facility. Paul Lomax is a global product manager for TTP Labtech, who are a UK based developer and supplier of automation solutions for the life science market including sample management, small volume liquid handling and detection systems.

Speaker
Biography:

Emi Hifumi has obtained her PhD from Kyushu University and studied Antibody Engineering and Catalytic Antibody. She was a Research Assistant at Hiroshima Prefectural University and is currently a Professor at Oita University from 2007. She has published more than 30 papers in reputed journals.

Abstract:

We are developing catalytic antibody light chains (human antigenases) by using the human genes belonging to subgroup II, which exhibit some unique features such as enzymatic function and also anti-virus infection. We amplified and cloned cDNAs encoding the human antibody light chains (kappa) belonging to subgroup II. The obtained cDNAs were transformed into E. coli and then expressed as the protein. The highly purified (over 95%) antigenases were submitted to the following experiments. Several antigenases out of over 200 antigenases investigated showed the suppressive effect on the infectivity of influenza virus H1N1 not only in vitro but also in vivo assay. 22F6 antigenase clearly prevented from the infection of influenza virus H1N1 in vivo, where PR-8 strain was used. The serum titer of the mice inoculated with antigenase treated virus was substantially low even at 21 dpi, comparing with the positive control, suggesting the lost of antigenicity of the virus. Taking into account that the antigenase showed the catalytic activity as DNase and RNase, the loss of the infectivity may be due to the cleavage of the virus RNA by the antigenase. On the other hand, 23D4m also possessed a suppressive function for the infection of influenza virus in vivo, while it is a monomeric light chain. In the investigation of nasal inoculation schedule of 23D4m, it clearly showed anti-viral effect under the simultaneous inoculation of the virus and the light chain. These results suggest that the above antigenases have the high possibility to prevent from the infection of influenza virus.

Speaker
Biography:

Enrica Pessione has completed her PhD in Microbiology at “Institut Pasteur”, Paris. She has been a Researcher in Microbial Biochemistry at Turin University getting expertise in enzymology, metabolic biochemistry and microbial proteomics. Since 2004, she is an Associate Professor in Biochemistry and Member of the Italian Microbial Biotechnologies Association. She has focused her interest on metabolic pathways important for food safety control or to establish the probiotic potential of lactic acid bacteria. She has published over 80 scientific articles. She is in the Editorial Board of Journal of Integrated Omics.

Abstract:

Lactic acid bacteria (LAB) have extensively been used as well-defined starters in the industry of fermented food production. More recently, their role as biocontrol agents to counteract food-borne infections and spoilage bacteria has also been proposed, to lower the amount of salts, sugar and preservatives in food, but also to reduce the cold-chain need, with benefits for costs and sustainability. Nowadays, LAB potential as probiotics and nutraceutical vectors has a huge impact on the pharmaceutical and food supplement industry. Although it is very promising to use LAB as microbial cell factories for functional food production, however, their metabolic profiles are often underexplored and most products underexploited. Proteomics is a promising tool for obtaining information on the metabolic pathways of interest and how these routes can be modulated by exogenous conditions. This presentation will refer all the cutting-edge products that can be obtained by LAB, the use of comparative proteomics and sub-proteome techniques to ascertain probiotic safety and efficacy, nutraceutical compounds release and optimization of the industrial production. In particular, selenium fixing ability, neuroactive compounds production, food-encrypted peptides release, antimicrobial molecules and exopolysaccharide (EPS) biosynthesis as well as bacterial resistance technological treatments will be considered.

Mohit Kapoor

University Health Network, Canada

Title: MicroRNAs: Potential biomarkers and therapeutic targets in osteoarthritis

Time : 16:00-16:30

Speaker
Biography:

Mohit Kapoor is a Senior Scientist at the Krembil Research Institute, University Health Network in Canada. He leads the Cartilage Biology and Joint Restorative Medicine Research within the Arthritis Program at the University Health Network. He has published over 60 research article, reviews and book chapters in high impact journals. He is a recipient of several prestigious Research and Fellowship Awards from various research organizations across the globe. He is also an Editor of book on osteoarthritis that was recently published in 2015 and sits on Review Panel of various research funding organizations.

Abstract:

Osteoarthritis (OA) is the most common form of arthritis and ageing-related joint pathology associated with degradation of the articular cartilage, synovial inflammation/fibrosis, subchondral bone remodeling and osteophyte formation. This disease results from alterations in the joint tissues that lead to pain, loss of motion and instability. The economic burden associated with this disease is substantial. Specific mechanisms associated with the joint destruction and associated pain during OA is largely unknown. Due to the lack of biomarkers, it is impossible to identify patients exhibiting early stages of OA, leading to severe joint destruction. Furthermore, due to poor understanding of the underlying disease mechanisms, no disease-modifying therapies to treat OA exist. MicroRNAs (miRNAs) are small non-coding RNAs that are expressed as primary stem loop precursors and undergo maturation by enzymatic processes. It is estimated that miRNAs regulate more than 60% of all coding genes and play pivotal roles in pathophysiological processes; including cell proliferation, differentiation, genomic stability, metabolism, apoptosis and aging. In this lecture, I will talk about how we employed a combination of gene expression analyses, computational biology and in vitro biological functional assays to identify panel of microRNAs as potential biomarkers and therapeutic targets in osteoarthritis.

Speaker
Biography:

Arata Katayama has completed his PhD from Tokyo Institute of Technology and worked as Postdoctoral fellow at University of California, Institute of Toxic Substances Research. Presently, he teaches Sanitary Engineering for undergraduate course and Ecotoxicology for graduate course as a Professor of Department of Civil and Environmental Engineering, Nagoya University, Japan. He also serves many governmental committees on environmental pollution, polychlorinated biphenyl remediation and monitoring of environmental quality.

Abstract:

Microbial remediation is one of the promising technologies for treating contaminated environment, especially anaerobic technology for the aquifer and sediments. Anaerobic microbial remediation does not require the soil excavation and/or aeration of aquifer, therefore, it is less expensive and energy saving. However, microorganisms tend to have a narrow range of degradation spectrum of toxic substances, although the polluted sites often contain multiple toxic substances. To this shortcoming of microbial capacity of detoxification of multiple toxic substances, we have conducted the study on a synthetic microbial community to widen the microbial spectrum in the degradation of toxic substances. In this study, we have combined the halogenated aromatics-respiring anaerobe (Dehalobacter), halogenated aliphatics-respiring anaerobe (Geobacter), aromactics-oxidizing anaerobe (Desulfatiglans) and hydrogen-producing anaerobe (Clostridium). The synthetic community was also conducted using multiple enriched cultures including these anaerobes. The experiments suggested that the combination of different microbial strains/community is successfully carried out to widen the degrading spectrum. Toxic metabolites from one anaerobe, hydrogen sulfide from sulfate-reducing anaerobe, caused the inhibitory effect on other members in synthetic anaerobe community. In the case of synthetic anaerobic community applied to the plug-flow reactor, pentachlorophenol, the examined pollutant was completely mineralized. All the microbial strains distributed into the narrow upstream area in the reactor based on the detection of functional genes. These results suggested that the design and construction of synthetic anaerobic community would be useful and important for the successful bioremediation.

Speaker
Biography:

Sharon Mendel Williams joined Coventry University as a Lecturer in the School of Life Sciences in November 2014. She worked as a Post-doctorate Research Fellow in both Chemistry and Biology departments of Warwick University for 8 years. Her research focuses on the biophysics and biochemistry of proteins, and understanding the mechanisms of enzymes. She has a wide range of depth and experience in molecular biology, biochemistry, and chemistry. She is a member of the Royal Society of Chemistry and has been awarded a grant from the RSC research fund to accomplish the current research.

Abstract:

Lignin is an organic polymer found in the cell walls of plants. Lignin can be used to create biofuels, or as an organic hydrocarbon source for a large variety of chemicals and polymers. However, lignin is very robust and current industrial processes for using it are inefficient. Therefore, a useable biological process for degrading lignin would be of great benefit. Understanding the pathway of lignin degradation, and the byproducts, is essential in order to be able to exploit the use of micro-organisms. Furthermore, we can characterize novel bio-products obtained by enzymatic oxidation of lignin, which could have very interesting applications for industrial biotechnology. In the current project Dyp-type peroxidases from Gram-negative Pseudomonas fluorescens Pf-5 and recombinant Sphingobacterium MnSOD1 and MnSOD2 cloned into E. coli and were investigated. These are bacterial enzymes that are already known to degrade lignin. Different genetic mutations were introduced, and the resulting enzymes were characterized by using them with different lignin substrates. The reaction compounds were analysed by reverse phase HPLC/ GC-MS. The goal is to improve the effectiveness of the enzymes, increase the production of the enzymesand degrade the lignin into different and more useable compounds. Any of these goals would be of valuable scientific and commercial benefit.

Betty Lee

United States Department of Commerce, USA

Title: Biosecurity and how export controls impact the life sciences

Time : 17:30-18:00

Speaker
Biography:

Betty Lee has a PhD from Dartmouth Medical School, USA, MS in Clinical Chemistry from the University of Windsor, Canada and MS in Biochemistry from LSU Medical Center, USA. She completed her Post-doctoral training at the National Institutes of Health, USA. She currently works as a Licensing Officer with the US Government. She educates industries and academia about the Export Administration Regulations (EAR) and participates in outreach. In addition, she participated in the policy review of the Executive Order titled “Optimizing the Security of Biological Select Agents and Toxins in the United States” signed by President Obama on July 2, 2010.

Abstract:

Balancing biosecurity and legitimate life science research has become a priority in recent years. The rise of biotechnology and informatics has made rapid advances in the 21st century. Such a convergence of biology and technology increases the pace of biological findings and the emergence of new technologies. Emerging technologies that could be used for biological warfare poses a formidable challenge because of the unpredictable nature of science. Dual use research in the life sciences requires some oversight by the government and funding agencies. The published results of scientific research could be used to improve health or agricultural products or they could be used to enable bioterrorism. Life Sciences research is conducted increasingly in an interdisciplinary and international environment. Informatics, systems biology, nanotechnology, and synthetic biology are at the forefront of such endeavors. Oversight of such research is essential to continue the free exchange of information and also balance the national security concerns. One of the tools that is at the nexus of biosecurity and life sciences is export control. The session will provide an overview of the biological agents (viruses, bacteria and toxins), genetic elements (DNA, plasmids, vectors) which are currently controlled on the Australia Group Control List and Commerce Control List for exports. The Australia Group is an informal forum of countries whichseeks to ensure that exports do not contribute to the development of chemical or biological weapons. Topics of discussion will include deemed exports, fundamental research and technology transfer.