Directed Evolution Charles Feng, Andrew Goodrich Team Presentation

Slides:



Advertisements
Similar presentations
Problem Results: Question: 1. You screen two libraries- cDNA; genomic
Advertisements

Recombinant DNA Technology
Systematic Evolution of Ligands by Exponential Enrichment: RNA Ligands to Bacteriophage T4 DNA Polymerase CRAIG TUERK AND LARRY GOLD.
Directed Evolution of a Fungal Peroxidase Irene Woo Enzong Yap Joel R. Cherry et al.
Darwinian Genomics Csaba Pal Biological Research Center Szeged, Hungary.
Putting biology to work for you: In vitro (directed) evolution and other techniques.
Key Area : Genetic Control of Metabolism in Micro-organisms Unit 2: Metabolism and Survival.
2 March, 2005 Chapter 12 Mutational dissection Normal gene Altered gene with altered phenotype mutagenesis.
Recombinant DNA Introduction to Recombinant DNA technology
Microbial Genetics As we have looked at, information for all cellular function is from DNA, mRNA carries that info to ribosomes, rRNA codes for proteins.
Lezione mercoledì 11 maggio 2011 corso vettori biologici II Biotec industriali ore 14:00 -16:00 aula 6A.
Physical Mapping I CIS 667 February 26, Physical Mapping A physical map of a piece of DNA tells us the location of certain markers  A marker is.
Evolutionary Computation Introduction Peter Andras s.
General Microbiology (Micr300) Lecture 10 Microbial Genetics (Text Chapter: ; )
CHAPTER 31 Genetic Engineering and Biotechnology.
1 Directed Mutagenesis and Protein Engineering. 2 Mutagenesis Mutagenesis -> change in DNA sequence -> Point mutations or large modifications Point mutations.
Mutagenesis Methods Lily Peterson April 5 th, 2010.
Study and engineering of gene function: mutagenesis I. Why mutagenize? II. Random mutagenesis, mutant selection schemes III. Site-directed mutagenesis,
THE BUILDING BLOCKS OF LIFE. BUILT FOR YOU Putting Engineering back into Protein Engineering Jun Liao, UC Santa Cruz Manfred K. Warmuth, UC Santa Cruz.
PLANT BIOTECHNOLOGY & GENETIC ENGINEERING (3 CREDIT HOURS)
General Microbiology (Micr300) Lecture 11 Biotechnology (Text Chapters: ; )
CHAPTER 10 Bacterial Genetics.
Manipulating the Genome: DNA Cloning and Analysis 20.1 – 20.3 Lesson 4.8.
DNA and Chromosome Structure. Chromosomal Structure of the Genetic Material.
An in vitro selection technique using a peptide or protein genetically fused to the coat protein of a bacteriophage.
Relationship between Genotype and Phenotype
Patenting Antibodies in Europe
Synthetic biology Genome engineering Chris Yellman, U. Texas CSSB.
DNA Technology and Genomics
Ch. 13 Genetic Engineering
Topic 6 Growth & Reproduction of Bacteria
Biotechnology: Bacterial Transformation Lab
AP Biology Ch. 20 Biotechnology.
Sequence analysis: what is a sequence? Linear arrangement of chemical subunits Contains information: 3-D arrangement determined by the sequence; 3-D defines.
Yeast as a Model System MBIOS 520/420 September 29, 2005.
Jonathan Sun University of Illinois at Urbana Champaign BIOE 506 February 15, 2010
Microbial Biotechnology Philadelphia University
Molecular Computing and Evolution In Vitro Evolution In Vitro Clustering Genetic Algorithms Artificial Immune Systems Biological Evolution and Molecular.
DNA Technology. Overview DNA technology makes it possible to clone genes for basic research and commercial applications DNA technology is a powerful set.
-Know that we can manipulate genomes by inserting or deleting certain genes. -What about synthesizing an entirely novel genome using sequencing technology?
PHARMACOBIOTECHNOLOGY.  Recombinant DNA (rDNA) is constructed outside the living cell using enzymes called “restriction enzymes” to cut DNA at specific.
Evolution of new protein topologies through multistep gene rearrangements Sergio G Peisajovich, Liat Rockah & Dan S Tawfik.
Chapter 16 Microbial Genomics “If we should succeed in helping ourselves through applied genetics before vengefully or accidentally exterminating ourselves,
Pairwise sequence alignment Lecture 02. Overview  Sequence comparison lies at the heart of bioinformatics analysis.  It is the first step towards structural.
Chapter 3 The Interrupted Gene.
Genetic Engineering/ Recombinant DNA Technology
What is phage display? An in vitro selection technique using a peptide or protein genetically fused to the coat protein of a bacteriophage.
Da-Hyeong Cho Protein Engineering Laboratory Department of Biotechnology and Bioengineering Sungkyunkwan University Site-Directed Mutagenesis.
Cloning of PCR Fragment into T- Vector Jung-Min Choi Department of Biochemistry, College of Life Science and Biotechnology, Mouse Genetics and Laboratory.
Directed Evolution of a Genetic Circuit 15 February 2008 George McArthur This presentation is primarily based on content taken from the paper “Directed.
Chapter 5 Evolution M13.  M13.1 Know that DNA IS UNIVERSAL TO MOST LIVING THINGS  DNA is the fundamental chemical of all living things  All living.
Biotechnology.
Biocatalysis, Dr. Rebecca Buller
Biotechnology and Recombinant DNA
Biotechnology and Recombinant DNA
Directed Mutagenesis and Protein Engineering
PCR TECHNIQUE
Chapter 4 Recombinant DNA Technology
Unit Genetic Control of Metabolism
Directed Mutagenesis and Protein Engineering
APPLICATIONS OF BIOINFORMATICS IN DRUG DISCOVERY
Additional DNA Technology AP Biology Ms. Day
Recombinant DNA Technology
Biotechnology and Recombinant DNA
Biotechnology - Theory and Application
Recombinant DNA Technology
Protein Engineering Protein engineering Industrial enzymes (Table 8.1)
Chapter 4 The Interrupted Gene.
Directed Mutagenesis and Protein Engineering
Biosynthetic phage display: a novel protein engineering tool combining chemical and genetic diversity  Mary A Dwyer, Wuyuan Lu, John J Dwyer, Anthony.
Presentation transcript:

Directed Evolution Charles Feng, Andrew Goodrich Team Presentation BIOE 506 Cellular & Molecular Bioengineering

The Issue At Hand Biotechnology requires specifically designed catalytic processes One option is biocatalytic processes using enzymes, but there’s only so many available Biocatalyst optimization has been a major topic, but we have limited predictive power for the relationship between structure and function for proteins So far, engineering of biocatalysts has been difficult and time-consuming

The Magic of Evolution All of nature’s complexity/beauty can be attributed to the “blind watchmaker” Mutation and its impact on life as a basis for natural selection Proteins as most basic element, function affects compatibility with environment Why can’t we do things the same way?

Protein Design Original ideas: forcing design on existing proteins, “top-down” approach More recently: directed evolution Buchholtz et al: improve function of site- specific FLP recombinase Kumamaru et al: polychlorinated biphenyl- degrading enzymes with novel substrates What’s so great about the above?

Differences between Lab/Natural Evolution Lab evolution is a “guided” process towards a final goal that may or may not make biological sense Natural evolution is a gradual accumulation of changes based on environmental factors

Major Challenge We’re not sure what affects performance and specificity! Thermostability? Activity? Solubility? Binding properties? Structure? Proteins too complex to manually change, as we don’t know effects of one change on other functions/behaviors Improving stability might adversely affect catalytic activity, etc.

The Solution Directed evolution lets proteins reinvent themselves, thereby eliminating the need for mindless tinkering Requirements: Function must be physically feasible Function must be biologically feasible Must be able to make libraries of mutants via a complex enough microorganism Must have a rapid screen or selection to evaluate the desired function

Screening for Function Need to combine two things: In vitro transcription/translation apparatus SIngle genes Tawfik and Griffiths: Combine in reverse micelles, select by evaluating modification of gene by its protein product Many other ideas out there

The Evolutionary Process More difficult problem - how do we force something to change in the way we want? Random mutagenesis - Arnold et al Can create enzyme variants on scale of months/weeks/days by rounds of mutagenesis and screening Family shuffling - Stemmer et al Homologous recombination of evolutionarily related genes Library of “chimeric genes” created that should fold in the same way as their precursors, but now there’s variation present

Mathematical Standpoint All possible changes/variations in amino acid sequence creates a multidimensional “performance landscape” We’re trying to go from one (biologically, naturally evolved) maximum to another that may be a distance away In order to get from one to the other, we need to use evolutionary strategies that take us along a stepwise variational path

Random Mutagenesis Error-prone PCR: method of choice if starting from single protein sequence Mutation rate is 1/2 mutations per protein so all variants can be exhaustively evaluated - more mutations would create combinatorial challenges Many created enzymes will be non/dysfunctional, evaluated through large screening libraries Promising/improved variants subsequently subjected to additional rounds of mutagenesis

Results of Mutagenesis Can successfully improve stability or activity of an enzyme - many specific solutions exist and mutations in iterative rounds are very additive Drawback - genetic code is conservative, many similar codons code for same amino acid or another amino acid w/ same properties

Homologous Recombination Alternatively we can use recombination to create chimeras of many homologous genes Advantages: will result in mostly functional variants b/c genes have already been naturally selected Can possibly create new functions Most common method: “family shuffling” - example is chimeric protein made from 6 parent sequences, now having 87-fold higher antiviral activity

Homologous Recombination Recombination works well for similar sequences Another study: 26 subtilisin sequences with 56.4% sequence identity Wide range of enzymatic properties including those not found in the parent Much better performance than parental gene Interesting point: sequence-wise, many times the best parent is dissimilar to best chimera suggesting that sequence isn’t everything Limitation of method: demands high sequence identity (normally 70%), difficulty of some crossover events based on parent gene sequence

RACHITT Developed by Coco et al to improve recombination efficiency Hybridize random DNA fragments to a single-stranded DNA scaffold, then trim overlaps, fill gaps, ligate nicks Subsequent digestion of resulting ds DNA strand can create chimeric DNA fragments Average 14 crossovers/gene variant versus 1-4 in previous shuffling techniques Allows for crossovers in dissimilar areas, i.e. those with less than five consecutive matching bases Technically more demanding

Nonhomologous Recombination Creation of fused enzyme libraries ITCHY: library of chimeric E. coli and human GAR (glycinamide ribonucleotide) as model system Ligation of truncated fragments from each organism Low frequency of functional chimeras Fusion occurred near central region of proteins SHIPREC: “sequence homology-independent protein recombination” Two genes truncated at restriction sites, then linearized and fragments cloned Correct reading frame established by adding chloramphenicol resistance gene in frame

Applications to Enzymes Enzyme stability and activity Good targets for directed evolution Additive mutations can lead to much improved variants Important for biocatalytic application Must be stable under both evolution process and application conditions Wintrode et al: low-temperature activity and high-temperature stability can be evolved independently

Applications to Enzymes Substrate specificity: Improving catalytic activity for new substrates Example: in vitro evolution of an aspartate aminotransferase with 1 million-fold increased efficiency for catalysis of non-native substrate valine Best chimeras have modified active sites (i.e. having contributions from both parents) P450 monooxygenases: promising for biotransformation applications - eight positions identified defining length of substrate it can act on

Applications to Enzymes Enantioselectivity Cofactor/activator requirements Resistance to oxidizing conditions Resistance to chemical modifications

Application to Binding Proteins Improving binding affinity to specific substrates, or binding capabilities to additional substrates Knappik et al: 40-fold higher antibody affinity for bovine insulin Stability of poorly folding anti-fluorescein binding antibody improved by grafting binding loops into better human antibody - further improved with mutagenesis

Creation of New Metabolic Pathways Modification/combination of existing pathways by evolving metabolic genes Can help with discovery of new, useful compounds TIM barrel fold protein: important protein found in many enzyme families catalyzing different reactions Transplant new catalytic activity on scaffold with existing binding site Transplant new binding site on scaffold with existing catalytic activity

Creation of New Metabolic Pathways New pathways for production of novel carotenoids Combine carotenoid biosynthetic genes from different microorganisms

Conclusions Directed evolution has potential for solving many bioenzymatic design problems: Improve enzyme substrate specificity, stability, activity, etc Improve protein binding affinity Create novel metabolic pathways In the future: applications to pathways, viruses, even complete genomes

Questions?