Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Cell ; 185(9): 1487-1505.e14, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35366417

ABSTRACT

Small molecules encoded by biosynthetic pathways mediate cross-species interactions and harbor untapped potential, which has provided valuable compounds for medicine and biotechnology. Since studying biosynthetic gene clusters in their native context is often difficult, alternative efforts rely on heterologous expression, which is limited by host-specific metabolic capacity and regulation. Here, we describe a computational-experimental technology to redesign genes and their regulatory regions with hybrid elements for cross-species expression in Gram-negative and -positive bacteria and eukaryotes, decoupling biosynthetic capacity from host-range constraints to activate silenced pathways. These synthetic genetic elements enabled the discovery of a class of microbiome-derived nucleotide metabolites-tyrocitabines-from Lactobacillus iners. Tyrocitabines feature a remarkable orthoester-phosphate, inhibit translational activity, and invoke unexpected biosynthetic machinery, including a class of "Amadori synthases" and "abortive" tRNA synthetases. Our approach establishes a general strategy for the redesign, expression, mobilization, and characterization of genetic elements in diverse organisms and communities.


Subject(s)
Biosynthetic Pathways , Host Microbial Interactions , Microbiota , Synthetic Biology/methods , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Genetic Engineering , Humans , Metabolomics
2.
Cell ; 171(6): 1453-1467.e13, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29153834

ABSTRACT

We describe a multiplex genome engineering technology in Saccharomyces cerevisiae based on annealing synthetic oligonucleotides at the lagging strand of DNA replication. The mechanism is independent of Rad51-directed homologous recombination and avoids the creation of double-strand DNA breaks, enabling precise chromosome modifications at single base-pair resolution with an efficiency of >40%, without unintended mutagenic changes at the targeted genetic loci. We observed the simultaneous incorporation of up to 12 oligonucleotides with as many as 60 targeted mutations in one transformation. Iterative transformations of a complex pool of oligonucleotides rapidly produced large combinatorial genomic diversity >105. This method was used to diversify a heterologous ß-carotene biosynthetic pathway that produced genetic variants with precise mutations in promoters, genes, and terminators, leading to altered carotenoid levels. Our approach of engineering the conserved processes of DNA replication, repair, and recombination could be automated and establishes a general strategy for multiplex combinatorial genome engineering in eukaryotes.


Subject(s)
Genetic Engineering/methods , Saccharomyces cerevisiae/genetics , DNA Replication , Escherichia coli/genetics , Gene Editing , Oligonucleotides/chemistry
3.
Nucleic Acids Res ; 51(22): 12522-12536, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37941137

ABSTRACT

The widespread Pseudomonas genus comprises a collection of related species with remarkable abilities to degrade plastics and polluted wastes and to produce a broad set of valuable compounds, ranging from bulk chemicals to pharmaceuticals. Pseudomonas possess characteristics of tolerance and stress resistance making them valuable hosts for industrial and environmental biotechnology. However, efficient and high-throughput genetic engineering tools have limited metabolic engineering efforts and applications. To improve their genome editing capabilities, we first employed a computational biology workflow to generate a genus-specific library of potential single-stranded DNA-annealing proteins (SSAPs). Assessment of the library was performed in different Pseudomonas using a high-throughput pooled recombinase screen followed by Oxford Nanopore NGS analysis. Among different active variants with variable levels of allelic replacement frequency (ARF), efficient SSAPs were found and characterized for mediating recombineering in the four tested species. New variants yielded higher ARFs than existing ones in Pseudomonas putida and Pseudomonas aeruginosa, and expanded the field of recombineering in Pseudomonas taiwanensisand Pseudomonas fluorescens. These findings will enhance the mutagenesis capabilities of these members of the Pseudomonas genus, increasing the possibilities for biotransformation and enhancing their potential for synthetic biology applications. .


Subject(s)
Gene Editing , Pseudomonas , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Gene Editing/methods , Metabolic Engineering , Pseudomonas/genetics , Pseudomonas putida/genetics
4.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046019

ABSTRACT

The use of biologics in the treatment of numerous diseases has increased steadily over the past decade due to their high specificities, low toxicity, and limited side effects. Despite this success, peptide- and protein-based drugs are limited by short half-lives and immunogenicity. To address these challenges, we use a genomically recoded organism to produce genetically encoded elastin-like polypeptide-protein fusions containing multiple instances of para-azidophenylalanine (pAzF). Precise lipidation of these pAzF residues generated a set of sequence-defined synthetic biopolymers with programmable binding affinity to albumin without ablating the activity of model fusion proteins, and with tunable blood serum half-lives spanning 5 to 94% of albumin's half-life in a mouse model. Our findings present a proof of concept for the use of genetically encoded bioorthogonal conjugation sites for multisite lipidation to tune protein stability in mouse serum. This work establishes a programmable approach to extend and tune the half-life of protein or peptide therapeutics and a technical foundation to produce functionalized biopolymers endowed with programmable chemical and biophysical properties with broad applications in medicine, materials science, and biotechnology.


Subject(s)
Biopolymers/chemistry , Lipids/chemistry , Peptides/chemistry , Proteins/chemistry , Amino Acids , Animals , Half-Life , Mice , Protein Engineering/methods , Synthetic Biology/methods
5.
Nat Chem Biol ; 17(6): 724-731, 2021 06.
Article in English | MEDLINE | ID: mdl-33820990

ABSTRACT

Genetically modified microorganisms (GMMs) can enable a wide range of important applications including environmental sensing and responsive engineered living materials. However, containment of GMMs to prevent environmental escape and satisfy regulatory requirements is a bottleneck for real-world use. While current biochemical strategies restrict unwanted growth of GMMs in the environment, there is a need for deployable physical containment technologies to achieve redundant, multi-layered and robust containment. We developed a hydrogel-based encapsulation system that incorporates a biocompatible multilayer tough shell and an alginate-based core. This deployable physical containment strategy (DEPCOS) allows no detectable GMM escape, bacteria to be protected against environmental insults including antibiotics and low pH, controllable lifespan and easy retrieval of genomically recoded bacteria. To highlight the versatility of DEPCOS, we demonstrated that robustly encapsulated cells can execute useful functions, including performing cell-cell communication with other encapsulated bacteria and sensing heavy metals in water samples from the Charles River.


Subject(s)
Bacteria/drug effects , Hydrogels/pharmacology , Alginates/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Biocompatible Materials , Bioengineering , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Environmental Monitoring , Escherichia coli/drug effects , Escherichia coli/genetics , Heme/chemistry , Metals, Heavy/chemistry , Organisms, Genetically Modified , Quorum Sensing , Rivers , Water Pollutants/chemistry
6.
Nat Rev Genet ; 16(9): 501-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26260262

ABSTRACT

Next-generation DNA sequencing has revealed the complete genome sequences of numerous organisms, establishing a fundamental and growing understanding of genetic variation and phenotypic diversity. Engineering at the gene, network and whole-genome scale aims to introduce targeted genetic changes both to explore emergent phenotypes and to introduce new functionalities. Expansion of these approaches into massively parallel platforms establishes the ability to generate targeted genome modifications, elucidating causal links between genotype and phenotype, as well as the ability to design and reprogramme organisms. In this Review, we explore techniques and applications in genome engineering, outlining key advances and defining challenges.


Subject(s)
Genetic Engineering/methods , Genome , High-Throughput Nucleotide Sequencing/methods , Animals , Gene Targeting/methods , Genetic Variation , Genotype , Humans
7.
Nature ; 518(7537): 89-93, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25607356

ABSTRACT

Genetically modified organisms (GMOs) are increasingly used in research and industrial systems to produce high-value pharmaceuticals, fuels and chemicals. Genetic isolation and intrinsic biocontainment would provide essential biosafety measures to secure these closed systems and enable safe applications of GMOs in open systems, which include bioremediation and probiotics. Although safeguards have been designed to control cell growth by essential gene regulation, inducible toxin switches and engineered auxotrophies, these approaches are compromised by cross-feeding of essential metabolites, leaked expression of essential genes, or genetic mutations. Here we describe the construction of a series of genomically recoded organisms (GROs) whose growth is restricted by the expression of multiple essential genes that depend on exogenously supplied synthetic amino acids (sAAs). We introduced a Methanocaldococcus jannaschii tRNA:aminoacyl-tRNA synthetase pair into the chromosome of a GRO derived from Escherichia coli that lacks all TAG codons and release factor 1, endowing this organism with the orthogonal translational components to convert TAG into a dedicated sense codon for sAAs. Using multiplex automated genome engineering, we introduced in-frame TAG codons into 22 essential genes, linking their expression to the incorporation of synthetic phenylalanine-derived amino acids. Of the 60 sAA-dependent variants isolated, a notable strain harbouring three TAG codons in conserved functional residues of MurG, DnaA and SerS and containing targeted tRNA deletions maintained robust growth and exhibited undetectable escape frequencies upon culturing ∼10(11) cells on solid media for 7 days or in liquid media for 20 days. This is a significant improvement over existing biocontainment approaches. We constructed synthetic auxotrophs dependent on sAAs that were not rescued by cross-feeding in environmental growth assays. These auxotrophic GROs possess alternative genetic codes that impart genetic isolation by impeding horizontal gene transfer and now depend on the use of synthetic biochemical building blocks, advancing orthogonal barriers between engineered organisms and the environment.


Subject(s)
Amino Acids/chemical synthesis , Amino Acids/pharmacology , Containment of Biohazards/methods , Escherichia coli/drug effects , Escherichia coli/genetics , Microbial Viability/drug effects , Synthetic Biology/methods , Amino Acids/chemistry , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Catalytic Domain/genetics , Codon/genetics , Culture Media/chemistry , Culture Media/pharmacology , Environment , Escherichia coli/cytology , Escherichia coli/metabolism , Escherichia coli Proteins/biosynthesis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Evolution, Molecular , Gene Transfer, Horizontal/genetics , Genes, Essential/genetics , Genetic Code/genetics , Genetic Engineering/methods , Genome, Bacterial/genetics , Microbial Viability/genetics , Molecular Sequence Data , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/growth & development , Organisms, Genetically Modified/metabolism , Peptide Termination Factors/genetics , Phenylalanine/chemistry , Phenylalanine/metabolism , Protein Multimerization/genetics , RNA, Transfer/genetics
8.
Nano Lett ; 20(12): 8890-8896, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33164530

ABSTRACT

Fluorescence microscopy has been one of the most discovery-rich methods in biology. In the digital age, the discipline is becoming increasingly quantitative. Virtually all biological laboratories have access to fluorescence microscopes, but abilities to quantify biomolecule copy numbers are limited by the complexity and sophistication associated with current quantification methods. Here, we present DNA-origami-based fluorescence brightness standards for counting 5-300 copies of proteins in bacterial and mammalian cells, tagged with fluorescent proteins or membrane-permeable organic dyes. Compared to conventional quantification techniques, our brightness standards are robust, straightforward to use, and compatible with nearly all fluorescence imaging applications, thereby providing a practical and versatile tool to quantify biomolecules via fluorescence microscopy.


Subject(s)
DNA , Fluorescent Dyes , Animals , Microscopy, Fluorescence , Proteins
9.
Nano Lett ; 19(1): 247-254, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30540482

ABSTRACT

Polypeptides are promising carriers for chemotherapeutics: they have minimal toxicity, can be recombinantly synthesized with precise control over molecular weight, and enhance drug pharmacokinetics as self-assembled nanoparticles. Polypeptide-based systems also provide the ability to achieve active targeting with genetically encoded targeting ligands. While passive targeting promotes accumulation of nanocarriers in solid tumors, active targeting provides an additional layer of tunable control and widens the therapeutic window. However, fusion of most targeting proteins to polypeptide carriers exposes the limitations of this approach: the residues that are used for drug attachment are also promiscuously distributed on protein surfaces. We present here a universal methodology to solve this problem by the site-specific attachment of extrinsic moieties to polypeptide drug delivery systems without cross-reactivity to fused targeting domains. We incorporate an unnatural amino acid, p-acetylphenylalanine, to provide a biorthogonal ketone for attachment of doxorubicin in the presence of reactive amino acids in a nanobody-targeted, elastin-like polypeptide nanoparticle. These nanoparticles exhibit significantly greater cytotoxicity than nontargeted controls in multiple cancer cell lines.


Subject(s)
Drug Delivery Systems , Nanoparticles/chemistry , Neoplasms/drug therapy , Peptides/chemistry , Animals , Cell Line, Tumor , Doxorubicin/chemistry , Doxorubicin/pharmacology , Elastin/chemistry , Elastin/pharmacology , Humans , Ligands , Micelles , Nanoparticles/administration & dosage , Peptides/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/pharmacology
10.
Nat Methods ; 13(2): 177-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26689263

ABSTRACT

Genetic regulatory proteins inducible by small molecules are useful synthetic biology tools as sensors and switches. Bacterial allosteric transcription factors (aTFs) are a major class of regulatory proteins, but few aTFs have been redesigned to respond to new effectors beyond natural aTF-inducer pairs. Altering inducer specificity in these proteins is difficult because substitutions that affect inducer binding may also disrupt allostery. We engineered an aTF, the Escherichia coli lac repressor, LacI, to respond to one of four new inducer molecules: fucose, gentiobiose, lactitol and sucralose. Using computational protein design, single-residue saturation mutagenesis or random mutagenesis, along with multiplex assembly, we identified new variants comparable in specificity and induction to wild-type LacI with its inducer, isopropyl ß-D-1-thiogalactopyranoside (IPTG). The ability to create designer aTFs will enable applications including dynamic control of cell metabolism, cell biology and synthetic gene circuits.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/genetics , Genetic Engineering , Lac Repressors/genetics , Lac Repressors/metabolism , Allosteric Regulation , DNA, Bacterial/genetics , Disaccharides , Escherichia coli/genetics , Fucose , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Sucrose/analogs & derivatives , Sugar Alcohols
11.
Proc Natl Acad Sci U S A ; 113(38): E5588-97, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601680

ABSTRACT

The degeneracy of the genetic code allows nucleic acids to encode amino acid identity as well as noncoding information for gene regulation and genome maintenance. The rare arginine codons AGA and AGG (AGR) present a case study in codon choice, with AGRs encoding important transcriptional and translational properties distinct from the other synonymous alternatives (CGN). We created a strain of Escherichia coli with all 123 instances of AGR codons removed from all essential genes. We readily replaced 110 AGR codons with the synonymous CGU codons, but the remaining 13 "recalcitrant" AGRs required diversification to identify viable alternatives. Successful replacement codons tended to conserve local ribosomal binding site-like motifs and local mRNA secondary structure, sometimes at the expense of amino acid identity. Based on these observations, we empirically defined metrics for a multidimensional "safe replacement zone" (SRZ) within which alternative codons are more likely to be viable. To evaluate synonymous and nonsynonymous alternatives to essential AGRs further, we implemented a CRISPR/Cas9-based method to deplete a diversified population of a wild-type allele, allowing us to evaluate exhaustively the fitness impact of all 64 codon alternatives. Using this method, we confirmed the relevance of the SRZ by tracking codon fitness over time in 14 different genes, finding that codons that fall outside the SRZ are rapidly depleted from a growing population. Our unbiased and systematic strategy for identifying unpredicted design flaws in synthetic genomes and for elucidating rules governing codon choice will be crucial for designing genomes exhibiting radically altered genetic codes.


Subject(s)
Arginine/genetics , Escherichia coli/genetics , RNA, Messenger/genetics , Amino Acids/genetics , Codon/genetics , Genes, Essential/genetics , Genetic Code , Genome, Bacterial , Protein Biosynthesis/genetics , RNA, Messenger/biosynthesis
12.
Genome Res ; 24(3): 365-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24347612

ABSTRACT

RNA molecules transmit the information encoded in the genome and generally reflect its content. Adenosine-to-inosine (A-to-I) RNA editing by ADAR proteins converts a genomically encoded adenosine into inosine. It is known that most RNA editing in human takes place in the primate-specific Alu sequences, but the extent of this phenomenon and its effect on transcriptome diversity are not yet clear. Here, we analyzed large-scale RNA-seq data and detected ∼1.6 million editing sites. As detection sensitivity increases with sequencing coverage, we performed ultradeep sequencing of selected Alu sequences and showed that the scope of editing is much larger than anticipated. We found that virtually all adenosines within Alu repeats that form double-stranded RNA undergo A-to-I editing, although most sites exhibit editing at only low levels (<1%). Moreover, using high coverage sequencing, we observed editing of transcripts resulting from residual antisense expression, doubling the number of edited sites in the human genome. Based on bioinformatic analyses and deep targeted sequencing, we estimate that there are over 100 million human Alu RNA editing sites, located in the majority of human genes. These findings set the stage for exploring how this primate-specific massive diversification of the transcriptome is utilized.


Subject(s)
Adenosine/genetics , Alu Elements , Inosine/metabolism , Primates/genetics , RNA Editing , Animals , Base Sequence , Gene Expression Regulation , Genes , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Transcriptome
13.
Biotechnol Bioeng ; 114(5): 1074-1086, 2017 05.
Article in English | MEDLINE | ID: mdl-27987323

ABSTRACT

The ability to site-specifically incorporate non-canonical amino acids (ncAAs) into proteins has made possible the study of protein structure and function in fundamentally new ways, as well as the bio synthesis of unnatural polymers. However, the task of site-specifically incorporating multiple ncAAs into proteins with high purity and yield continues to present a challenge. At the heart of this challenge lies the lower efficiency of engineered orthogonal translation system components compared to their natural counterparts (e.g., translation elements that specifically use a ncAA and do not interact with the cell's natural translation apparatus). Here, we show that evolving and tuning expression levels of multiple components of an engineered translation system together as a whole enhances ncAA incorporation efficiency. Specifically, we increase protein yield when incorporating multiple p-azido-phenylalanine(pAzF) residues into proteins by (i) evolving the Methanocaldococcus jannaschii p-azido-phenylalanyl-tRNA synthetase anti-codon binding domain, (ii) evolving the elongation factor Tu amino acid-binding pocket, and (iii) tuning the expression of evolved translation machinery components in a single vector. Use of the evolved translation machinery in a genomically recoded organism lacking release factor one enabled enhanced multi-site ncAA incorporation into proteins. We anticipate that our approach to orthogonal translation system development will accelerate and expand our ability to site-specifically incorporate multiple ncAAs into proteins and biopolymers, advancing new horizons for synthetic and chemical biotechnology. Biotechnol. Bioeng. 2017;114: 1074-1086. © 2016 Wiley Periodicals, Inc.


Subject(s)
Amino Acids/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Protein Biosynthesis , Protein Engineering/methods , Amino Acyl-tRNA Synthetases/metabolism , Models, Molecular , Peptide Elongation Factor Tu/metabolism , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Proteins/genetics , Proteins/metabolism
14.
Nucleic Acids Res ; 43(3): 1945-54, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25567985

ABSTRACT

Genetically modified organisms (GMOs) are commonly used to produce valuable compounds in closed industrial systems. However, their emerging applications in open clinical or environmental settings require enhanced safety and security measures. Intrinsic biocontainment, the creation of bacterial hosts unable to survive in natural environments, remains a major unsolved biosafety problem. We developed a new biocontainment strategy containing overlapping 'safeguards'-engineered riboregulators that tightly control expression of essential genes, and an engineered addiction module based on nucleases that cleaves the host genome-to restrict viability of Escherichia coli cells to media containing exogenously supplied synthetic small molecules. These multilayered safeguards maintain robust growth in permissive conditions, eliminate persistence and limit escape frequencies to <1.3 × 10(-12). The staged approach to safeguard implementation revealed mechanisms of escape and enabled strategies to overcome them. Our safeguarding strategy is modular and employs conserved mechanisms that could be extended to clinically or industrially relevant organisms and undomesticated species.


Subject(s)
Escherichia coli/genetics , Organisms, Genetically Modified/growth & development , Cloning, Molecular , Coculture Techniques , Culture Media , Escherichia coli/growth & development , Recombination, Genetic
16.
Nucleic Acids Res ; 42(7): 4779-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24452804

ABSTRACT

Selection has been invaluable for genetic manipulation, although counter-selection has historically exhibited limited robustness and convenience. TolC, an outer membrane pore involved in transmembrane transport in E. coli, has been implemented as a selectable/counter-selectable marker, but counter-selection escape frequency using colicin E1 precludes using tolC for inefficient genetic manipulations and/or with large libraries. Here, we leveraged unbiased deep sequencing of 96 independent lineages exhibiting counter-selection escape to identify loss-of-function mutations, which offered mechanistic insight and guided strain engineering to reduce counter-selection escape frequency by ∼40-fold. We fundamentally improved the tolC counter-selection by supplementing a second agent, vancomycin, which reduces counter-selection escape by 425-fold, compared colicin E1 alone. Combining these improvements in a mismatch repair proficient strain reduced counter-selection escape frequency by 1.3E6-fold in total, making tolC counter-selection as effective as most selectable markers, and adding a valuable tool to the genome editing toolbox. These improvements permitted us to perform stable and continuous rounds of selection/counter-selection using tolC, enabling replacement of 10 alleles without requiring genotypic screening for the first time. Finally, we combined these advances to create an optimized E. coli strain for genome engineering that is ∼10-fold more efficient at achieving allelic diversity than previous best practices.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins/genetics , Genetic Engineering/methods , Membrane Transport Proteins/genetics , Alleles , Biomarkers , Escherichia coli/genetics , Gene Deletion , Gene Duplication , Genome, Bacterial , High-Throughput Nucleotide Sequencing , Phenotype
17.
Nature ; 460(7257): 894-898, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19633652

ABSTRACT

The breadth of genomic diversity found among organisms in nature allows populations to adapt to diverse environments. However, genomic diversity is difficult to generate in the laboratory and new phenotypes do not easily arise on practical timescales. Although in vitro and directed evolution methods have created genetic variants with usefully altered phenotypes, these methods are limited to laborious and serial manipulation of single genes and are not used for parallel and continuous directed evolution of gene networks or genomes. Here, we describe multiplex automated genome engineering (MAGE) for large-scale programming and evolution of cells. MAGE simultaneously targets many locations on the chromosome for modification in a single cell or across a population of cells, thus producing combinatorial genomic diversity. Because the process is cyclical and scalable, we constructed prototype devices that automate the MAGE technology to facilitate rapid and continuous generation of a diverse set of genetic changes (mismatches, insertions, deletions). We applied MAGE to optimize the 1-deoxy-D-xylulose-5-phosphate (DXP) biosynthesis pathway in Escherichia coli to overproduce the industrially important isoprenoid lycopene. Twenty-four genetic components in the DXP pathway were modified simultaneously using a complex pool of synthetic DNA, creating over 4.3 billion combinatorial genomic variants per day. We isolated variants with more than fivefold increase in lycopene production within 3 days, a significant improvement over existing metabolic engineering techniques. Our multiplex approach embraces engineering in the context of evolution by expediting the design and evolution of organisms with new and improved properties.


Subject(s)
Biotechnology/methods , Directed Molecular Evolution/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Genome, Bacterial/genetics , Alleles , Biotechnology/instrumentation , Carotenoids/biosynthesis , Chromosomes, Bacterial/genetics , DNA/biosynthesis , DNA/genetics , Directed Molecular Evolution/instrumentation , Escherichia coli/cytology , Genetic Variation/genetics , Lycopene , Pentosephosphates/biosynthesis
18.
J Biol Chem ; 288(17): 12295-304, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23486477

ABSTRACT

Antifreeze proteins (AFPs) help some organisms resist freezing by binding to ice crystals and inhibiting their growth. The molecular basis for how these proteins recognize and bind ice is not well understood. The longhorn beetle Rhagium inquisitor can supercool to below -25 °C, in part by synthesizing the most potent antifreeze protein studied thus far (RiAFP). We report the crystal structure of the 13-kDa RiAFP, determined at 1.21 Å resolution using direct methods. The structure, which contains 1,914 nonhydrogen protein atoms in the asymmetric unit, is the largest determined ab initio without heavy atoms. It reveals a compressed ß-solenoid fold in which the top and bottom sheets are held together by a silk-like interdigitation of short side chains. RiAFP is perhaps the most regular structure yet observed. It is a second independently evolved AFP type in beetles. The two beetle AFPs have in common an extremely flat ice-binding surface comprising regular outward-projecting parallel arrays of threonine residues. The more active, wider RiAFP has four (rather than two) of these arrays between which the crystal structure shows the presence of ice-like waters. Molecular dynamics simulations independently reproduce the locations of these ordered crystallographic waters and predict additional waters that together provide an extensive view of the AFP interaction with ice. By matching several planes of hexagonal ice, these waters may help freeze the AFP to the ice surface, thus providing the molecular basis of ice binding.


Subject(s)
Antifreeze Proteins/chemistry , Ice , Insect Proteins/chemistry , Molecular Dynamics Simulation , Protein Folding , Animals , Coleoptera , Crystallography, X-Ray , Protein Structure, Secondary
19.
Nucleic Acids Res ; 40(17): e132, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22638574

ABSTRACT

Genome-scale engineering of living organisms requires precise and economical methods to efficiently modify many loci within chromosomes. One such example is the directed integration of chemically synthesized single-stranded deoxyribonucleic acid (oligonucleotides) into the chromosome of Escherichia coli during replication. Herein, we present a general co-selection strategy in multiplex genome engineering that yields highly modified cells. We demonstrate that disparate sites throughout the genome can be easily modified simultaneously by leveraging selectable markers within 500 kb of the target sites. We apply this technique to the modification of 80 sites in the E. coli genome.


Subject(s)
Genetic Engineering/methods , Oligonucleotides/chemistry , Chromosomes, Bacterial , Escherichia coli/genetics , Genome, Bacterial
20.
Nat Commun ; 15(1): 5218, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890276

ABSTRACT

Technologies that generate precise combinatorial genome modifications are well suited to dissect the polygenic basis of complex phenotypes and engineer synthetic genomes. Genome modifications with engineered nucleases can lead to undesirable repair outcomes through imprecise homology-directed repair, requiring non-cleavable gene editing strategies. Eukaryotic multiplex genome engineering (eMAGE) generates precise combinatorial genome modifications in Saccharomyces cerevisiae without generating DNA breaks or using engineered nucleases. Here, we systematically optimize eMAGE to achieve 90% editing frequency, reduce workflow time, and extend editing distance to 20 kb. We further engineer an inducible dominant negative mismatch repair system, allowing for high-efficiency editing via eMAGE while suppressing the elevated background mutation rate 17-fold resulting from mismatch repair inactivation. We apply these advances to construct a library of cancer-associated mutations in the ligand-binding domains of human estrogen receptor alpha and progesterone receptor to understand their impact on ligand-independent autoactivation. We validate that this yeast model captures autoactivation mutations characterized in human breast cancer models and further leads to the discovery of several previously uncharacterized autoactivating mutations. This work demonstrates the development and optimization of a cleavage-free method of genome editing well suited for applications requiring efficient multiplex editing with minimal background mutations.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Mutation , Saccharomyces cerevisiae , Gene Editing/methods , Saccharomyces cerevisiae/genetics , Humans , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , DNA Mismatch Repair/genetics , Breast Neoplasms/genetics , Female
SELECTION OF CITATIONS
SEARCH DETAIL