Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Elife ; 122024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836839

ABSTRACT

New experimental findings continue to challenge our understanding of protein allostery. Recent deep mutational scanning study showed that allosteric hotspots in the tetracycline repressor (TetR) and its homologous transcriptional factors are broadly distributed rather than spanning well-defined structural pathways as often assumed. Moreover, hotspot mutation-induced allostery loss was rescued by distributed additional mutations in a degenerate fashion. Here, we develop a two-domain thermodynamic model for TetR, which readily rationalizes these intriguing observations. The model accurately captures the in vivo activities of various mutants with changes in physically transparent parameters, allowing the data-based quantification of mutational effects using statistical inference. Our analysis reveals the intrinsic connection of intra- and inter-domain properties for allosteric regulation and illustrate epistatic interactions that are consistent with structural features of the protein. The insights gained from this study into the nature of two-domain allostery are expected to have broader implications for other multi-domain allosteric proteins.


Subject(s)
Mutation , Repressor Proteins , Thermodynamics , Allosteric Regulation , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/genetics , Protein Domains , Models, Molecular
2.
Trends Genet ; 40(7): 587-600, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38658256

ABSTRACT

Population-scale sequencing efforts have catalogued substantial genetic variation in humans such that variant discovery dramatically outpaces interpretation. We discuss how single-cell sequencing is poised to reveal genetic mechanisms at a rate that may soon approach that of variant discovery. The functional genomics toolkit is sufficiently modular to systematically profile almost any type of variation within increasingly diverse contexts and with molecularly comprehensive and unbiased readouts. As a result, we can construct deep phenotypic atlases of variant effects that span the entire regulatory cascade. The same conceptual approach to interpreting genetic variation should be applied to engineering therapeutic cell states. In this way, variant mechanism discovery and cell state engineering will become reciprocating and iterative processes towards genomic medicine.


Subject(s)
Genetic Variation , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Genomics/methods , Genome, Human/genetics , Phenotype
3.
bioRxiv ; 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38659955

ABSTRACT

Bacterial host factors regulate the infection cycle of bacteriophages. Except for some well-studied host factors (e.g., receptors or restriction-modification systems), the contribution of the rest of the host genome on phage infection remains poorly understood. We developed PHAGEPACK, a pooled assay that systematically and comprehensively measures each host-gene impact on phage fitness. PHAGEPACK combines CRISPR interference with phage packaging to link host perturbation to phage fitness during active infection. Using PHAGEPACK, we constructed a genome-wide map of genes impacting T7 phage fitness in permissive E. coli, revealing pathways previously unknown to affect phage packaging. When applied to the non-permissive E. coli O121, PHAGEPACK identified pathways leading to host resistance; their removal increased phage susceptibility up to a billion-fold. Bioinformatic analysis indicates phage genomes carry homologs or truncations of key host factors, potentially for fitness advantage. In summary, PHAGEPACK offers valuable insights into phage-host interactions, phage evolution, and bacterial resistance.

4.
Cell Syst ; 15(4): 374-387.e6, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38537640

ABSTRACT

How a protein's function influences the shape of its fitness landscape, smooth or rugged, is a fundamental question in evolutionary biochemistry. Smooth landscapes arise when incremental mutational steps lead to a progressive change in function, as commonly seen in enzymes and binding proteins. On the other hand, rugged landscapes are poorly understood because of the inherent unpredictability of how sequence changes affect function. Here, we experimentally characterize the entire sequence phylogeny, comprising 1,158 extant and ancestral sequences, of the DNA-binding domain (DBD) of the LacI/GalR transcriptional repressor family. Our analysis revealed an extremely rugged landscape with rapid switching of specificity, even between adjacent nodes. Further, the ruggedness arises due to the necessity of the repressor to simultaneously evolve specificity for asymmetric operators and disfavors potentially adverse regulatory crosstalk. Our study provides fundamental insight into evolutionary, molecular, and biophysical rules of genetic regulation through the lens of fitness landscapes.


Subject(s)
Phylogeny
5.
bioRxiv ; 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38496486

ABSTRACT

Allosteric transcription factors (aTF), widely used as biosensors, have proven challenging to design for detecting novel molecules because mutation of ligand-binding residues often disrupts allostery. We developed Sensor-seq, a high-throughput platform to design and identify aTF biosensors that bind to non-native ligands. We screened a library of 17,737 variants of the aTF TtgR, a regulator of a multidrug exporter, against six non-native ligands of diverse chemical structures - four derivatives of the cancer therapeutic tamoxifen, the antimalarial drug quinine, and the opiate analog naltrexone - as well as two native flavonoid ligands, naringenin and phloretin. Sensor-seq identified novel biosensors for each of these ligands with high dynamic range and diverse specificity profiles. The structure of a naltrexone-bound design showed shape-complementary methionine-aromatic interactions driving ligand specificity. To demonstrate practical utility, we developed cell-free detection systems for naltrexone and quinine. Sensor-seq enables rapid, scalable design of new biosensors, overcoming constraints of natural biosensors.

6.
bioRxiv ; 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-37662419

ABSTRACT

New experimental findings continue to challenge our understanding of protein allostery. Recent deep mutational scanning study showed that allosteric hotspots in the tetracycline repressor (TetR) and its homologous transcriptional factors are broadly distributed rather than spanning well-defined structural pathways as often assumed. Moreover, hotspot mutation-induced allostery loss was rescued by distributed additional mutations in a degenerate fashion. Here, we develop a two-domain thermodynamic model for TetR, which readily rationalizes these intriguing observations. The model accurately captures the in vivo activities of various mutants with changes in physically transparent parameters, allowing the data-based quantification of mutational effects using statistical inference. Our analysis reveals the intrinsic connection of intra- and inter-domain properties for allosteric regulation and illustrate epistatic interactions that are consistent with structural features of the protein. The insights gained from this study into the nature of two-domain allostery are expected to have broader implications for other multidomain allosteric proteins.

7.
bioRxiv ; 2023 Sep 21.
Article in English | MEDLINE | ID: mdl-37781603

ABSTRACT

Although population-scale databases have expanded to millions of protein-coding variants, insight into variant mechanisms has not kept pace. We present PROD-ATAC, a high-throughput method for discovering the effects of protein-coding variants on chromatin. A pooled library of variants is expressed in a disease-agnostic cell line, and single-cell ATAC resolves each variant's effect on chromatin. Using PROD-ATAC, we characterized the effects of >100 oncofusions (a class of cancer-causing chimeric proteins) and controls and revealed that pioneer activity is a common feature of fusions spanning an enormous range of fusion frequencies. Further, fusion-induced dysregulation can be context-agnostic as observed mechanisms often overlapped with cancer and cell-type specific prior knowledge. We also showed that gain-of-function pioneering is common among oncofusions. This work provides a global view of fusion-induced chromatin. We uncovered convergent mechanisms among disparate oncofusions and shared modes of dysregulation across different cancers. PROD-ATAC is generalizable to any set of protein-coding variants.

8.
bioRxiv ; 2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36798209

ABSTRACT

Bacteriophages can adapt to new hosts by altering sequence motifs through recombination or convergent evolution. Where such motifs exist and what fitness advantage they confer remains largely unknown. We report a new method, Bacteriophage Library Informed Sequence Scoring (BLISS), to discover sequence motifs in metagenomic datasets governing phage activity. BLISS uses experimental deep mutational scanning data to create sequence profiles to enable deep mining of metagenomes for functional motifs which are otherwise invisible to searches. We experimentally tested 10,073 BLISS-derived sequence motifs for the receptor-binding protein of the T7 phage. The screen revealed hundreds of T7 variants with novel host specificity with functional motifs sourced from distant families besides other major phyla. Position, substitution and location preferences on T7 dictated different specificities. To demonstrate therapeutic utility, we engineered highly active T7 variants against urinary tract pathogens. BLISS is a powerful tool to unlock the functional potential encoded in phage metagenomes.

9.
Trends Biochem Sci ; 48(2): 187-197, 2023 02.
Article in English | MEDLINE | ID: mdl-36180320

ABSTRACT

Bacteriophage research has been vital to fundamental aspects of modern biology. Advances in metagenomics have revealed treasure troves of new and uncharacterized bacteriophages ('phages') that are not yet understood. However, our ability to find new phages has outpaced our understanding of how sequence encodes function in phages. Traditional approaches for characterizing phages are limited in scale and face hurdles in determining how changes in sequence drive function. We describe powerful emerging technologies that can be used to clarify sequence-function relationships in phages through high-throughput genome engineering. Using these approaches, up to 105 variants can be characterized through pooled selection experiments and deep sequencing. We describe caveats when using these tools and provide examples of basic science and engineering goals that are pursuable using these approaches.


Subject(s)
Bacteriophages , Bacteriophages/genetics , Genome, Viral
10.
Elife ; 112022 10 13.
Article in English | MEDLINE | ID: mdl-36226916

ABSTRACT

A fundamental question in protein science is where allosteric hotspots - residues critical for allosteric signaling - are located, and what properties differentiate them. We carried out deep mutational scanning (DMS) of four homologous bacterial allosteric transcription factors (aTFs) to identify hotspots and built a machine learning model with this data to glean the structural and molecular properties of allosteric hotspots. We found hotspots to be distributed protein-wide rather than being restricted to 'pathways' linking allosteric and active sites as is commonly assumed. Despite structural homology, the location of hotspots was not superimposable across the aTFs. However, common signatures emerged when comparing hotspots coincident with long-range interactions, suggesting that the allosteric mechanism is conserved among the homologs despite differences in molecular details. Machine learning with our large DMS datasets revealed global structural and dynamic properties to be a strong predictor of whether a residue is a hotspot than local and physicochemical properties. Furthermore, a model trained on one protein can predict hotspots in a homolog. In summary, the overall allosteric mechanism is embedded in the structural fold of the aTF family, but the finer, molecular details are sequence-specific.


Subject(s)
Machine Learning , Proteins , Allosteric Regulation , Mutation , Proteins/chemistry , Signal Transduction , Molecular Dynamics Simulation
11.
ACS Synth Biol ; 11(1): 286-296, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34985866

ABSTRACT

Transcriptional repressors play an important role in regulating phage life cycle. Here, we examine how synthetic transcription repressors can be used in bacteriophage T7 to create a dynamic, controllable infectivity switch. We engineered T7 phage by replacing a large region of the early phage genome with different combinations of ligand-responsive promoters and ribosome binding sites (RBS) designed to control the phage RNA polymerase, gp1. Phages with engineered infectivity switch are fully viable at levels comparable to wildtype T7, when not repressed, indicating the phage can be engineered without loss of fitness. The most effective switch used a TetR-responsive promoter and an attenuated RBS, resulting in a 2-fold increase in latent period and a 10-fold decrease in phage titer when repressed. Phage activity can be further tuned using different inducer concentrations. Our study provides a proof of concept for how a simple synthetic circuit introduced into the phage genome enables user control over phage infectivity.


Subject(s)
Bacteriophage T7 , Bacteriophages , Bacteriophage T7/genetics , Bacteriophages/genetics , DNA-Directed RNA Polymerases/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics
12.
Nat Commun ; 12(1): 5562, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34548494

ABSTRACT

Epistasis is a major determinant in the emergence of novel protein function. In allosteric proteins, direct interactions between inducer-binding mutations propagate through the allosteric network, manifesting as epistasis at the level of biological function. Elucidating this relationship between local interactions and their global effects is essential to understanding evolution of allosteric proteins. We integrate computational design, structural and biophysical analysis to characterize the emergence of novel inducer specificity in an allosteric transcription factor. Adaptive landscapes of different inducers of the designed mutant show that a few strong epistatic interactions constrain the number of viable sequence pathways, revealing ridges in the fitness landscape leading to new specificity. The structure of the designed mutant shows that a striking change in inducer orientation still retains allosteric function. Comparing biophysical and functional properties suggests a nonlinear relationship between inducer binding affinity and allostery. Our results highlight the functional and evolutionary complexity of allosteric proteins.


Subject(s)
Allosteric Regulation/genetics , Epistasis, Genetic , Genetic Fitness/genetics , Transcription Factors/genetics , Allosteric Site , Computer Simulation , Crystallography, X-Ray , Evolution, Molecular , Ligands , Models, Genetic , Mutation , Protein Binding , Transcription Factors/chemistry , Transcription Factors/metabolism
13.
Cell Rep ; 36(5): 109471, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348151

ABSTRACT

Viruses influence the fate of nutrients and human health by killing microorganisms and altering metabolic processes. Organosulfur metabolism and biologically derived hydrogen sulfide play dynamic roles in manifestation of diseases, infrastructure degradation, and essential biological processes. Although microbial organosulfur metabolism is well studied, the role of viruses in organosulfur metabolism is unknown. Here, we report the discovery of 39 gene families involved in organosulfur metabolism encoded by 3,749 viruses from diverse ecosystems, including human microbiomes. The viruses infect organisms from all three domains of life. Six gene families encode for enzymes that degrade organosulfur compounds into sulfide, whereas others manipulate organosulfur compounds and may influence sulfide production. We show that viral metabolic genes encode key enzymatic domains, are translated into protein, and are maintained after recombination, and sulfide provides a fitness advantage to viruses. Our results reveal viruses as drivers of organosulfur metabolism with important implications for human and environmental health.


Subject(s)
Environment , Organic Chemicals/metabolism , Sulfur/metabolism , Viruses/metabolism , Gastrointestinal Microbiome , Genes, Viral , Genetic Variation , Genomics , Humans , Metabolic Networks and Pathways/genetics , Microbiota , Phylogeny , Recombination, Genetic/genetics , Sulfides/metabolism , Viruses/genetics
14.
Elife ; 102021 03 09.
Article in English | MEDLINE | ID: mdl-33687327

ABSTRACT

The interaction between a bacteriophage and its host is mediated by the phage's receptor binding protein (RBP). Despite its fundamental role in governing phage activity and host range, molecular rules of RBP function remain a mystery. Here, we systematically dissect the functional role of every residue in the tip domain of T7 phage RBP (1660 variants) by developing a high-throughput, locus-specific, phage engineering method. This rich dataset allowed us to cross compare functional profiles across hosts to precisely identify regions of functional importance, many of which were previously unknown. Substitution patterns showed host-specific differences in position and physicochemical properties of mutations, revealing molecular adaptation to individual hosts. We discovered gain-of-function variants against resistant hosts and host-constricting variants that eliminated certain hosts. To demonstrate therapeutic utility, we engineered highly active T7 variants against a urinary tract pathogen. Our approach presents a generalized framework for characterizing sequence-function relationships in many phage-bacterial systems.


Bacteria can cause diseases, but they also battle their own microscopic enemies: a group of viruses known as bacteriophages. For instance, the T7 bacteriophage preys on various strains of Escherichia coli, a type of bacteria often found in the human gut. While many E. coli strains are inoffensive or even beneficial to human health, some can be deadly. Finding a way to kill harmful strains while sparing the helpful ones would be a helpful addition to the medicine toolkit. Bacteriophages identify and interact with their specific target through a structure known as the receptor binding protein, or RBP. However, it is still unclear exactly how RBP helps the viruses recognize which type of bacteria to infect. Here, Huss et al. set to map out and modify this structure in T7 bacteriophage so the virus is more efficient and specific about which strain of E. coli it kills. First, the role of each building block in the tip of RBP was meticulously dissected; this generated the knowledge required to genetically engineer a large number of different T7 bacteriophages, each with a slightly variation in their RBP. These viruses were then exposed to various strains of bacteria. Monitoring the bacteriophages that survived and multiplied the most after infecting different strains of E. coli revealed which RBP building blocks are important for efficiency and specificity. This was then confirmed by engineering highly active T7 bacteriophage variants against an E. coli strain that causes urinary tract infections. These findings demonstrate that even small changes to the bacteriophages can make a big difference to their ability to infect their preys. The approaches developed by Huss et al. help to understand exactly how the RBP allows a virus to infect a specific type of bacteria; this could one day pave the way for new therapies that harness those viruses to fight increasingly resistant bacterial infections.


Subject(s)
Bacteriophage T7/genetics , Mutation , Genetic Techniques , Protein Binding , Viral Proteins
15.
Nat Chem Biol ; 17(5): 531-539, 2021 05.
Article in English | MEDLINE | ID: mdl-33526893

ABSTRACT

Splitting bioactive proteins into conditionally reconstituting fragments is a powerful strategy for building tools to study and control biological systems. However, split proteins often exhibit a high propensity to reconstitute, even without the conditional trigger, limiting their utility. Current approaches for tuning reconstitution propensity are laborious, context-specific or often ineffective. Here, we report a computational design strategy grounded in fundamental protein biophysics to guide experimental evaluation of a sparse set of mutants to identify an optimal functional window. We hypothesized that testing a limited set of mutants would direct subsequent mutagenesis efforts by predicting desirable mutant combinations from a vast mutational landscape. This strategy varies the degree of interfacial destabilization while preserving stability and catalytic activity. We validate our method by solving two distinct split protein design challenges, generating both design and mechanistic insights. This new technology will streamline the generation and use of split protein systems for diverse applications.


Subject(s)
Molecular Probes/chemistry , Protein Engineering/methods , Transcription Factors/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Genes, Reporter , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Probes/genetics , Molecular Probes/metabolism , Mutation , Protein Multimerization , Proteolysis , Sirolimus/metabolism , Sirolimus/pharmacology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation
16.
Proc Natl Acad Sci U S A ; 117(41): 25445-25454, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32999067

ABSTRACT

Allostery is a fundamental regulatory mechanism of protein function. Despite notable advances, understanding the molecular determinants of allostery remains an elusive goal. Our current knowledge of allostery is principally shaped by a structure-centric view, which makes it difficult to understand the decentralized character of allostery. We present a function-centric approach using deep mutational scanning to elucidate the molecular basis and underlying functional landscape of allostery. We show that allosteric signaling exhibits a high degree of functional plasticity and redundancy through myriad mutational pathways. Residues critical for allosteric signaling are surprisingly poorly conserved while those required for structural integrity are highly conserved, suggesting evolutionary pressure to preserve fold over function. Our results suggest multiple solutions to the thermodynamic conditions of cooperativity, in contrast to the common view of a finely tuned allosteric residue network maintained under selection.


Subject(s)
Adaptation, Physiological , Allosteric Regulation/genetics , Bacteria/cytology , Bacterial Physiological Phenomena , Biological Evolution , Cloning, Molecular , Epigenesis, Genetic , Flow Cytometry , Gene Expression Regulation, Enzymologic , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Conformation
17.
ACS Synth Biol ; 9(1): 84-94, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31825601

ABSTRACT

Rapid molecular biosensing is an emerging application area for synthetic biology. Here, we engineer a portable biosensor for cyanuric acid (CYA), an analyte of interest for human and environmental health, using a LysR-type transcription regulator (LTTR) from Pseudomonas within the context of Escherichia coli gene expression machinery. To overcome cross-host portability challenges of LTTRs, we rationally engineered hybrid Pseudomonas-E. coli promoters by integrating DNA elements required for transcriptional activity and ligand-dependent regulation from both hosts, which enabled E. coli to function as a whole-cell biosensor for CYA. To alleviate challenges of whole-cell biosensing, we adapted these promoter designs to function within a freeze-dried E. coli cell-free system to sense CYA. This portable, on-demand system robustly detects CYA within an hour from laboratory and real-world samples and works with both fluorescent and colorimetric reporters. This work elucidates general principles to facilitate the engineering of a wider array of LTTR-based environmental sensors.


Subject(s)
Biosensing Techniques/methods , Escherichia coli/genetics , Pseudomonas/genetics , Transcription, Genetic , Triazines/analysis , Bacterial Proteins/metabolism , Cell-Free System , Chimera/genetics , Gene Expression , Gene Expression Regulation, Bacterial , Ligands , Plasmids/genetics , Promoter Regions, Genetic , Synthetic Biology/methods , Transcription Factors/metabolism
18.
Curr Opin Biotechnol ; 61: 116-121, 2020 02.
Article in English | MEDLINE | ID: mdl-31862543

ABSTRACT

Bacteriophages (or 'phages') can be potent biocontrol agents but their potential has not been fully realized due to inherent limitations of natural phages. By leveraging new tools in synthetic biology, natural phages can be engineered to overcome these limitations to markedly improve their efficacy and programmability. Engineered phages can be used for targeted detection and removal of pathogens, in situ microbiome editing, gene delivery and programmable control of phage-bacterial interactions. In this mini review we examine different ways natural phages can be engineered as effective biocontrol agents through a design-build-test-learn platform and identify novel applications of engineered phages in food biotechnology.


Subject(s)
Bacteriophages , Bacteria , Synthetic Biology
19.
Nucleic Acids Res ; 47(19): 10452-10463, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31552424

ABSTRACT

Ligand-responsive allosteric transcription factors (aTF) play a vital role in genetic circuits and high-throughput screening because they transduce biochemical signals into gene expression changes. Programmable control of gene expression from aTF-regulated promoter is important because different downstream effector genes function optimally at different expression levels. However, tuning gene expression of native promoters is difficult due to complex layers of homeostatic regulation encoded within them. We engineered synthetic promoters de novo by embedding operator sites with varying affinities and radically reshaped binding preferences within a minimal, constitutive Escherichia coli promoter. Multiplexed cell-based screening of promoters for three TetR-like aTFs generated with this approach gave rich diversity of gene expression levels, dynamic ranges and ligand sensitivities and were 50- to 100-fold more active over their respective native promoters. Machine learning on our dataset revealed that relative position of the core motif and bases flanking the core motif play an important role in modulating induction response. Our generalized approach yields customizable and programmable aTF-regulated promoters for engineering cellular pathways and enables the discovery of new small molecule biosensors.


Subject(s)
Allosteric Regulation/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/biosynthesis , Transcription, Genetic , Escherichia coli/genetics , Gene Expression Regulation/genetics , Ligands , Metabolic Engineering , Synthetic Biology , Transcription Factors/genetics
20.
ACS Synth Biol ; 8(2): 264-273, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30633862

ABSTRACT

NADH and NAD+ cofactors drive hundreds of biochemical reactions, and their ratio is a key metabolic marker of cellular state. Traditional assays to measure the NADH/NAD+ ratio is laborious, prone to inaccuracies, and not suitable for high-throughput screening. We report a genetically encoded ratiometric biosensor for NADH/NAD+ based on redox-responsive bacterial transcription factor Rex that overcomes these limitations. We engineered a Rex-regulated E. coli promoter with improved biosensor characteristics by tuning the affinity of Rex and the operator site. Since NADH is oxidized during aerobic respiration, we used the biosensor-reporter to investigate the effect of removing respiratory chain enzymes on NADH/NAD+ ratio during aerobiosis. We found that the NADH/NAD+ signal increased in five of the nine mutants by over 3-fold compared to wildtype, including an NADH dehydrogenase double mutant with 6-fold elevation. We also found that among several common carbon sources, E. coli grown on acetate exhibited higher NADH/NAD+ compared to E. coli grown on glucose. As a proof-of-concept for high-throughput redox screening, we were able to enrich high NADH mutants present at 1 in 10 000 among wildtype cells by biosensor-guided pooled screen. Thus, our Rex biosensor-reporter enables facile, noninvasive, high-throughput redox measurement to understand and engineer redox metabolism.


Subject(s)
Biosensing Techniques/methods , NAD/metabolism , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/metabolism , Oxidation-Reduction , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...