Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 910136, 2022.
Article in English | MEDLINE | ID: mdl-35911728

ABSTRACT

We assessed if immune responses are enhanced in CD-1 mice by heterologous vaccination with two different nucleic acid-based COVID-19 vaccines: a next-generation human adenovirus serotype 5 (hAd5)-vectored dual-antigen spike (S) and nucleocapsid (N) vaccine (AdS+N) and a self-amplifying and -adjuvanted S RNA vaccine (AAHI-SC2) delivered by a nanostructured lipid carrier. The AdS+N vaccine encodes S modified with a fusion motif to increase cell-surface expression and an N antigen modified with an Enhanced T-cell Stimulation Domain (N-ETSD) to direct N to the endosomal/lysosomal compartment and increase MHC class I and II stimulation potential. The S sequence in the AAHI-SC2 vaccine comprises the D614G mutation, two prolines to stabilize S in the prefusion conformation, and 3 glutamines in the furin cleavage region to confer protease resistance. CD-1 mice received vaccination by homologous and heterologous prime > boost combinations. Humoral responses to S were the highest with any regimen that included the AAHI-SC2 vaccine, and IgG bound to wild type and Delta (B.1.617.2) variant S1 at similar levels. An AAHI-SC2 prime followed by an AdS+N boost particularly enhanced CD4+ and CD8+ T-cell responses to both wild type and Delta S peptides relative to all other vaccine regimens. Sera from mice receiving AAHI-SC2 homologous or heterologous vaccination were found to be highly neutralizing for all pseudovirus strains tested: Wuhan, Beta, Delta, and Omicron strains. The findings here, taken in consideration with the availability of both vaccines in thermostable formulations, support the testing of heterologous vaccination by an AAHI-SC2 > AdS+N regimen in animal models of SARS-CoV-2 infection to assess its potential to provide increased protection against emerging SARS-CoV-2 variants particularly in regions of the world where the need for cold-chain storage has limited the distribution of other vaccines.


Subject(s)
COVID-19 , Viral Vaccines , Animals , Antibodies, Neutralizing , Antigens, Heterophile , COVID-19/prevention & control , COVID-19 Vaccines , DNA , Humans , Mice , SARS-CoV-2 , Vaccination , Vaccines, Synthetic , mRNA Vaccines
2.
Cell Rep ; 38(6): 110348, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35114110

ABSTRACT

The increasing prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with the ability to escape existing humoral protection conferred by previous infection and/or immunization necessitates the discovery of broadly reactive neutralizing antibodies (nAbs). Utilizing mRNA display, we identify a set of antibodies against SARS-CoV-2 spike (S) proteins and characterize the structures of nAbs that recognize epitopes in the S1 subunit of the S glycoprotein. These structural studies reveal distinct binding modes for several antibodies, including the targeting of rare cryptic epitopes in the receptor-binding domain (RBD) of S that interact with angiotensin-converting enzyme 2 (ACE2) to initiate infection, as well as the S1 subdomain 1. Further, we engineer a potent ACE2-blocking nAb to sustain binding to S RBD with the E484K and L452R substitutions found in multiple SARS-CoV-2 variants. We demonstrate that mRNA display is an approach for the rapid identification of nAbs that can be used in combination to combat emerging SARS-CoV-2 variants.

3.
Sci Rep ; 11(1): 12740, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140558

ABSTRACT

The SARS-CoV-2 variants replacing the first wave strain pose an increased threat by their potential ability to escape pre-existing humoral protection. An angiotensin converting enzyme 2 (ACE2) decoy that competes with endogenous ACE2 for binding of the SARS-CoV-2 spike receptor binding domain (S RBD) and inhibits infection may offer a therapeutic option with sustained efficacy against variants. Here, we used Molecular Dynamics (MD) simulation to predict ACE2 sequence substitutions that might increase its affinity for S RBD and screened candidate ACE2 decoys in vitro. The lead ACE2(T27Y/H34A)-IgG1FC fusion protein with enhanced S RBD affinity shows greater live SARS-CoV-2 virus neutralization capability than wild type ACE2. MD simulation was used to predict the effects of S RBD variant mutations on decoy affinity that was then confirmed by testing of an ACE2 Triple Decoy that included an additional enzyme activity-deactivating H374N substitution against mutated S RBD. The ACE2 Triple Decoy maintains high affinity for mutated S RBD, displays enhanced affinity for S RBD N501Y or L452R, and has the highest affinity for S RBD with both E484K and N501Y mutations, making it a viable therapeutic option for the prevention or treatment of SARS-CoV-2 infection with a high likelihood of efficacy against variants.


Subject(s)
Amino Acid Substitution , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , COVID-19/metabolism , Drug Discovery/methods , Molecular Dynamics Simulation , SARS-CoV-2/metabolism , Signal Transduction/drug effects , Amino Acid Sequence , COVID-19/virology , Humans , Mutation , Protein Binding/drug effects , Protein Domains/genetics , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects
5.
Proc Natl Acad Sci U S A ; 114(8): 2018-2023, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28159892

ABSTRACT

Hepatitis C virus (HCV) encodes mechanisms to evade the multilayered antiviral actions of the host immune system. Great progress has been made in elucidating the strategies HCV employs to down-regulate interferon (IFN) production, impede IFN signaling transduction, and impair IFN-stimulated gene (ISG) expression. However, there is a limited understanding of the mechanisms governing how viral proteins counteract the antiviral functions of downstream IFN effectors due to the lack of an efficient approach to identify such interactions systematically. To study the mechanisms by which HCV antagonizes the IFN responses, we have developed a high-throughput profiling platform that enables mapping of HCV sequences critical for anti-IFN function at high resolution. Genome-wide profiling performed with a 15-nt insertion mutant library of HCV showed that mutations in the p7 region conferred high levels of IFN sensitivity, which could be alleviated by the expression of WT p7 protein. This finding suggests that p7 protein of HCV has an immune evasion function. By screening a liver-specific ISG library, we identified that IFI6-16 significantly inhibits the replication of p7 mutant viruses without affecting WT virus replication. In contrast, knockout of IFI6-16 reversed the IFN hypersensitivity of p7 mutant virus. In addition, p7 was found to be coimmunoprecipitated with IFI6-16 and to counteract the function of IFI6-16 by depolarizing the mitochondria potential. Our data suggest that p7 is a critical immune evasion protein that suppresses the antiviral IFN function by counteracting the function of IFI6-16.


Subject(s)
Hepacivirus/pathogenicity , Hepatitis C/immunology , Host-Pathogen Interactions/immunology , Immune Evasion , Interferons/immunology , Mitochondrial Proteins/immunology , Viral Proteins/immunology , CRISPR-Cas Systems , Cell Line , Gene Expression Profiling , Gene Knockout Techniques , Gene Library , Genome, Viral , Hepacivirus/genetics , Hepatitis C/virology , Humans , Immunity, Innate , Interferons/genetics , Interferons/metabolism , Liver/immunology , Liver/metabolism , Membrane Potential, Mitochondrial/immunology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutagenesis, Insertional , Signal Transduction , Viral Proteins/genetics , Virus Replication
6.
Elife ; 52016 07 08.
Article in English | MEDLINE | ID: mdl-27391790

ABSTRACT

The structure of fitness landscapes is critical for understanding adaptive protein evolution. Previous empirical studies on fitness landscapes were confined to either the neighborhood around the wild type sequence, involving mostly single and double mutants, or a combinatorially complete subgraph involving only two amino acids at each site. In reality, the dimensionality of protein sequence space is higher (20(L)) and there may be higher-order interactions among more than two sites. Here we experimentally characterized the fitness landscape of four sites in protein GB1, containing 20(4) = 160,000 variants. We found that while reciprocal sign epistasis blocked many direct paths of adaptation, such evolutionary traps could be circumvented by indirect paths through genotype space involving gain and subsequent loss of mutations. These indirect paths alleviate the constraint on adaptive protein evolution, suggesting that the heretofore neglected dimensions of sequence space may change our views on how proteins evolve.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Evolution, Molecular , Mutation
7.
Protein Sci ; 25(2): 530-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26540565

ABSTRACT

The effect of a mutation on protein stability is traditionally measured by genetic construction, expression, purification, and physical analysis using low-throughput methods. This process is tedious and limits the number of mutants able to be examined in a single study. In contrast, functional fitness effects can be measured in a high-throughput manner by various deep mutational scanning tools. Using protein GB 1, we have recently demonstrated the feasibility of estimating the mutational stability effect ( ΔΔG) of single-substitution based on the functional fitness profile of all double-substitutions. The principle is to identify genetic backgrounds that have an exhausted stability margin. The functional effect of an additional substitution on these genetic backgrounds can then be used to compute the mutational ΔΔG based on the biophysical relationship between functional fitness and thermodynamic stability. However, to identify such genetic backgrounds, the approach described in our previous study required a benchmark dataset, which is a set of known mutational ΔΔG. In this study, a benchmark-independent approach is developed. The genetic backgrounds of interest are identified using k-means clustering with the integration of structural information. We further demonstrated that a reasonable approximation of ΔΔG can also be obtained without taking structural information into account. In summary, this study describes a novel method for computing ΔΔG from double-substitution functional fitness profiles alone, without relying on any known mutational ΔΔG as a benchmark.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Mutation , Streptococcus/chemistry , Streptococcus/genetics , Amino Acid Substitution , Models, Molecular , Protein Stability , Protein Structure, Tertiary , Thermodynamics
8.
PLoS Genet ; 11(7): e1005310, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132554

ABSTRACT

Viruses often encode proteins with multiple functions due to their compact genomes. Existing approaches to identify functional residues largely rely on sequence conservation analysis. Inferring functional residues from sequence conservation can produce false positives, in which the conserved residues are functionally silent, or false negatives, where functional residues are not identified since they are species-specific and therefore non-conserved. Furthermore, the tedious process of constructing and analyzing individual mutations limits the number of residues that can be examined in a single study. Here, we developed a systematic approach to identify the functional residues of a viral protein by coupling experimental fitness profiling with protein stability prediction using the influenza virus polymerase PA subunit as the target protein. We identified a significant number of functional residues that were influenza type-specific and were evolutionarily non-conserved among different influenza types. Our results indicate that type-specific functional residues are prevalent and may not otherwise be identified by sequence conservation analysis alone. More importantly, this technique can be adapted to any viral (and potentially non-viral) protein where structural information is available.


Subject(s)
Influenza A virus/genetics , Influenza B virus/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Base Sequence , Biological Evolution , Cell Line , Computational Biology , Conserved Sequence/genetics , Gene Library , HEK293 Cells , Humans , Sequence Analysis, DNA
9.
Retrovirology ; 11: 124, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25522661

ABSTRACT

BACKGROUND: The HIV-1 pandemic is not the result of a static pathogen but a large genetically diverse and dynamic viral population. The virus is characterized by a highly mutable genome rendering efforts to design a universal vaccine a significant challenge and drives the emergence of drug resistant variants upon antiviral pressure. Gaining a comprehensive understanding of the mutational tolerance of each HIV-1 genomic position is therefore of critical importance. RESULTS: Here we combine high-density mutagenesis with the power of next-generation sequencing to gauge the replication capacity and therefore mutational tolerability of single point mutations across the entire HIV-1 genome. We were able to achieve the evaluation of point mutational effects on viral replicative capacity for 5,553 individual HIV-1 nucleotide positions - representing 57% of the viral genome. Replicative capacity was assessed at 3,943 nucleotide positions for a single alternate base change, 1,459 nucleotide positions for two alternate base changes, and 151 nucleotide positions for all three possible alternate base changes. This resulted in the study of how a total of 7,314 individual point mutations impact HIV-1 replication on a single experimental platform. We further utilize the dataset for a focused structural analysis on a capsid inhibitor binding pocket. CONCLUSION: The approach presented here can be applied to any pathogen that can be genetically manipulated in a laboratory setting. Furthermore, the methodology can be utilized under externally applied selection conditions, such as drug or immune pressure, to identify genetic elements that contribute to drug or host interactions, and therefore mutational routes of pathogen resistance and escape.


Subject(s)
DNA Mutational Analysis/methods , Genome, Viral , HIV-1/genetics , Point Mutation , HIV-1/physiology , High-Throughput Nucleotide Sequencing , Humans , Molecular Biology/methods , Mutagenesis , Virology/methods , Virus Replication
10.
Curr Biol ; 24(22): 2643-51, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25455030

ABSTRACT

BACKGROUND: Nonadditivity in fitness effects from two or more mutations, termed epistasis, can result in compensation of deleterious mutations or negation of beneficial mutations. Recent evidence shows the importance of epistasis in individual evolutionary pathways. However, an unresolved question in molecular evolution is how often and how significantly fitness effects change in alternative genetic backgrounds. RESULTS: To answer this question, we quantified the effects of all single mutations and double mutations between all positions in the IgG-binding domain of protein G (GB1). By observing the first two steps of all possible evolutionary pathways using this fitness profile, we were able to characterize the extent and magnitude of pairwise epistasis throughout an entire protein molecule. Furthermore, we developed a novel approach to quantitatively determine the effects of single mutations on structural stability (ΔΔGU). This enabled determination of the importance of stability effects in functional epistasis. CONCLUSIONS: Our results illustrate common biophysical mechanisms for occurrences of positive and negative epistasis. Our results show pervasive positive epistasis within a conformationally dynamic network of residues. The stability analysis shows that significant negative epistasis, which is more common than positive epistasis, mostly occurs between combinations of destabilizing mutations. Furthermore, we show that although significant positive epistasis is rare, many deleterious mutations are beneficial in at least one alternative mutational background. The distribution of conditionally beneficial mutations throughout the domain demonstrates that the functional portion of sequence space can be significantly expanded by epistasis.


Subject(s)
Bacterial Proteins/chemistry , Epistasis, Genetic , Evolution, Molecular , Models, Genetic , Bacterial Proteins/genetics , Models, Molecular , Mutation , Protein Stability , Protein Structure, Tertiary
11.
mBio ; 5(5): e01469-14, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25271282

ABSTRACT

UNLABELLED: Pairing high-throughput sequencing technologies with high-throughput mutagenesis enables genome-wide investigations of pathogenic organisms. Knowledge of the specific functions of protein domains encoded by the genome of the hepatitis C virus (HCV), a major human pathogen that contributes to liver disease worldwide, remains limited to insight from small-scale studies. To enhance the capabilities of HCV researchers, we have obtained a high-resolution functional map of the entire viral genome by combining transposon-based insertional mutagenesis with next-generation sequencing. We generated a library of 8,398 mutagenized HCV clones, each containing one 15-nucleotide sequence inserted at a unique genomic position. We passaged this library in hepatic cells, recovered virus pools, and simultaneously assayed the abundance of mutant viruses in each pool by next-generation sequencing. To illustrate the validity of the functional profile, we compared the genetic footprints of viral proteins with previously solved protein structures. Moreover, we show the utility of these genetic footprints in the identification of candidate regions for epitope tag insertion. In a second application, we screened the genetic footprints for phenotypes that reflected defects in later steps of the viral life cycle. We confirmed that viruses with insertions in a region of the nonstructural protein NS4B had a defect in infectivity while maintaining genome replication. Overall, our genome-wide HCV mutant library and the genetic footprints obtained by high-resolution profiling represent valuable new resources for the research community that can direct the attention of investigators toward unidentified roles of individual protein domains. IMPORTANCE: Our insertional mutagenesis library provides a resource that illustrates the effects of relatively small insertions on local protein structure and HCV viability. We have also generated complementary resources, including a website (http://hangfei.bol.ucla.edu) and a panel of epitope-tagged mutant viruses that should enhance the research capabilities of investigators studying HCV. Researchers can now detect epitope-tagged viral proteins by established antibodies, which will allow biochemical studies of HCV proteins for which antibodies are not readily available. Furthermore, researchers can now quickly look up genotype-phenotype relationships and base further mechanistic studies on the residue-by-residue information from the functional profile. More broadly, this approach offers a general strategy for the systematic functional characterization of viruses on the genome scale.


Subject(s)
Genome, Viral , Hepacivirus/genetics , Viral Proteins/genetics , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA Transposable Elements/genetics , DNA, Viral/genetics , Gene Library , Hepacivirus/physiology , High-Throughput Nucleotide Sequencing , Humans , Mutagenesis, Insertional , Plasmids , Sequence Analysis, DNA , Transcription, Genetic , Transfection , Viral Proteins/metabolism , Virus Replication
12.
J Virol ; 88(17): 10157-64, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965464

ABSTRACT

UNLABELLED: Viral proteins often display several functions which require multiple assays to dissect their genetic basis. Here, we describe a systematic approach to screen for loss-of-function mutations that confer a fitness disadvantage under a specified growth condition. Our methodology was achieved by genetically monitoring a mutant library under two growth conditions, with and without interferon, by deep sequencing. We employed a molecular tagging technique to distinguish true mutations from sequencing error. This approach enabled us to identify mutations that were negatively selected against, in addition to those that were positively selected for. Using this technique, we identified loss-of-function mutations in the influenza A virus NS segment that were sensitive to type I interferon in a high-throughput fashion. Mechanistic characterization further showed that a single substitution, D92Y, resulted in the inability of NS to inhibit RIG-I ubiquitination. The approach described in this study can be applied under any specified condition for any virus that can be genetically manipulated. IMPORTANCE: Traditional genetics focuses on a single genotype-phenotype relationship, whereas high-throughput genetics permits phenotypic characterization of numerous mutants in parallel. High-throughput genetics often involves monitoring of a mutant library with deep sequencing. However, deep sequencing suffers from a high error rate (∼0.1 to 1%), which is usually higher than the occurrence frequency for individual point mutations within a mutant library. Therefore, only mutations that confer a fitness advantage can be identified with confidence due to an enrichment in the occurrence frequency. In contrast, it is impossible to identify deleterious mutations using most next-generation sequencing techniques. In this study, we have applied a molecular tagging technique to distinguish true mutations from sequencing errors. It enabled us to identify mutations that underwent negative selection, in addition to mutations that experienced positive selection. This study provides a proof of concept by screening for loss-of-function mutations on the influenza A virus NS segment that are involved in its anti-interferon activity.


Subject(s)
Influenza A virus/immunology , Influenza A virus/physiology , Interferon Type I/antagonists & inhibitors , Mutation , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/metabolism , High-Throughput Nucleotide Sequencing , Influenza A virus/genetics , Influenza A virus/growth & development , Molecular Biology/methods , RNA, Viral/genetics , Virology/methods
13.
Sci Rep ; 4: 4942, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24820965

ABSTRACT

Genetic research on influenza virus biology has been informed in large part by nucleotide variants present in seasonal or pandemic samples, or individual mutants generated in the laboratory, leaving a substantial part of the genome uncharacterized. Here, we have developed a single-nucleotide resolution genetic approach to interrogate the fitness effect of point mutations in 98% of the amino acid positions in the influenza A virus hemagglutinin (HA) gene. Our HA fitness map provides a reference to identify indispensable regions to aid in drug and vaccine design as targeting these regions will increase the genetic barrier for the emergence of escape mutations. This study offers a new platform for studying genome dynamics, structure-function relationships, virus-host interactions, and can further rational drug and vaccine design. Our approach can also be applied to any virus that can be genetically manipulated.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , High-Throughput Nucleotide Sequencing , Influenza A Virus, H1N1 Subtype/genetics , Polymorphism, Single Nucleotide , Binding Sites , Cell Line , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Models, Molecular , Mutation , Phenotype , Protein Conformation , Protein Interaction Domains and Motifs , Structure-Activity Relationship
14.
PLoS One ; 9(5): e97505, 2014.
Article in English | MEDLINE | ID: mdl-24842159

ABSTRACT

Trade-offs between throughput, read length, and error rates in high-throughput sequencing limit certain applications such as monitoring viral quasispecies. Here, we describe a molecular-based tag linkage method that allows assemblage of short sequence reads into long DNA fragments. It enables haplotype phasing with high accuracy and sensitivity to interrogate individual viral sequences in a quasispecies. This approach is demonstrated to deduce ∼ 2000 unique 1.3 kb viral sequences from HIV-1 quasispecies in vivo and after passaging ex vivo with a detection limit of ∼ 0.005% to ∼ 0.001%. Reproducibility of the method is validated quantitatively and qualitatively by a technical replicate. This approach can improve monitoring of the genetic architecture and evolution dynamics in any quasispecies population.


Subject(s)
HIV-1/genetics , High-Throughput Nucleotide Sequencing/methods , Genome, Viral/genetics , Reproducibility of Results
15.
PLoS Pathog ; 10(4): e1004064, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722365

ABSTRACT

Widely used chemical genetic screens have greatly facilitated the identification of many antiviral agents. However, the regions of interaction and inhibitory mechanisms of many therapeutic candidates have yet to be elucidated. Previous chemical screens identified Daclatasvir (BMS-790052) as a potent nonstructural protein 5A (NS5A) inhibitor for Hepatitis C virus (HCV) infection with an unclear inhibitory mechanism. Here we have developed a quantitative high-resolution genetic (qHRG) approach to systematically map the drug-protein interactions between Daclatasvir and NS5A and profile genetic barriers to Daclatasvir resistance. We implemented saturation mutagenesis in combination with next-generation sequencing technology to systematically quantify the effect of every possible amino acid substitution in the drug-targeted region (domain IA of NS5A) on replication fitness and sensitivity to Daclatasvir. This enabled determination of the residues governing drug-protein interactions. The relative fitness and drug sensitivity profiles also provide a comprehensive reference of the genetic barriers for all possible single amino acid changes during viral evolution, which we utilized to predict clinical outcomes using mathematical models. We envision that this high-resolution profiling methodology will be useful for next-generation drug development to select drugs with higher fitness costs to resistance, and also for informing the rational use of drugs based on viral variant spectra from patients.


Subject(s)
Drug Resistance, Viral , Gene Expression Profiling , Genetic Fitness , Hepacivirus/physiology , Hepatitis C , Imidazoles/pharmacology , Virus Replication , Carbamates , Cell Line , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Hepatitis C/drug therapy , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , Pyrrolidines , Valine/analogs & derivatives , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Virus Replication/genetics
16.
J Antimicrob Chemother ; 69(3): 724-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24169581

ABSTRACT

OBJECTIVES: Daclatasvir is a highly potent inhibitor of hepatitis C virus. We estimated the active tissue concentration of daclatasvir in vivo. METHODS: We developed a mathematical model incorporating pharmacokinetic/pharmacodynamic and viral dynamics. By fitting the model to clinical data reported previously, we estimated the ratio between plasma drug concentration and active tissue concentration in vivo. RESULTS: The modelling results show that the active tissue concentration of daclatasvir is ∼9% of the concentration measured in plasma (95% CI 1%-29%). CONCLUSIONS: Using plasma concentrations as surrogates for clinical recommendations may lead to substantial underestimation of the risk of resistance.


Subject(s)
Antiviral Agents/pharmacokinetics , Hepacivirus/drug effects , Imidazoles/pharmacokinetics , Plasma/chemistry , Antiviral Agents/administration & dosage , Carbamates , Clinical Trials as Topic , Hepacivirus/isolation & purification , Humans , Imidazoles/administration & dosage , Models, Theoretical , Pyrrolidines , Valine/analogs & derivatives , Viral Load
17.
Neuron ; 78(6): 971-85, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23791193

ABSTRACT

The ability to visualize endogenous proteins in living neurons provides a powerful means to interrogate neuronal structure and function. Here we generate recombinant antibody-like proteins, termed Fibronectin intrabodies generated with mRNA display (FingRs), that bind endogenous neuronal proteins PSD-95 and Gephyrin with high affinity and that, when fused to GFP, allow excitatory and inhibitory synapses to be visualized in living neurons. Design of the FingR incorporates a transcriptional regulation system that ties FingR expression to the level of the target and reduces background fluorescence. In dissociated neurons and brain slices, FingRs generated against PSD-95 and Gephyrin did not affect the expression patterns of their endogenous target proteins or the number or strength of synapses. Together, our data indicate that PSD-95 and Gephyrin FingRs can report the localization and amount of endogenous synaptic proteins in living neurons and thus may be used to study changes in synaptic strength in vivo.


Subject(s)
Carrier Proteins/analysis , Gene Expression Profiling/methods , Intracellular Signaling Peptides and Proteins/analysis , Membrane Proteins/analysis , Neurons/chemistry , Recombinant Proteins/analysis , Animals , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , Disks Large Homolog 4 Protein , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Neurons/physiology , Recombinant Proteins/genetics , Synapses/chemistry , Synapses/physiology
18.
Angew Chem Int Ed Engl ; 51(50): 12449-53, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23125174

ABSTRACT

In a single round: By combining the high-efficiency enrichment through the continuous-flow magnetic separation (CFMS) technique with the analytical power of next-generation sequencing, the generation of antibody mimetics with a single round of mRNA display is made possible. This approach eliminates iterative selection cycles and provides a path to fully automated ligand generation (see picture).


Subject(s)
Biomimetic Materials/metabolism , RNA, Messenger/chemistry , Amino Acid Sequence , Antibodies/chemistry , Antibodies/metabolism , Biomimetic Materials/chemistry , Enzyme-Linked Immunosorbent Assay , Fibronectins/chemistry , Fibronectins/metabolism , Gene Library , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/metabolism , Immunomagnetic Separation , Ligands , Maltose-Binding Proteins/chemistry , Maltose-Binding Proteins/metabolism , RNA, Messenger/isolation & purification
20.
ACS Nano ; 4(11): 6914-22, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21028792

ABSTRACT

Biosensors utilizing carbon nanotube field-effect transistors have a tremendous potential to serve as the basis for the next generation of diagnostic systems. While nanotubes have been employed in the fabrication of multiple sensors, little attention has previously been paid to how the nanotube density affects the biosensor performance. We conducted a systematic study of the effect of density on the performance of nanotube biosensors and discovered that this parameter is crucial to achieving consistently high performance. We found that devices with lower density offer higher sensitivity in terms of both detection limit and magnitude of response. The low density nanotube devices resulted in a detection limit of 1 pM in an electrolyte buffer containing high levels of electrolytes (ionic concentration ∼140 mM, matching the ionic strength of serum and plasma). Further investigation suggested that the enhanced sensitivity arises from the semiconductor-like behavior-strong gate dependence and lower capacitance-of the nanotube network at low density. Finally, we used the density-optimized nanotube biosensors to detect the nucleocapsid (N) protein of the SARS virus and demonstrated improved detection limits under physiological conditions. Our results show that it is critical to carefully tune the nanotube density in order to fabricate sensitive and reliable devices.


Subject(s)
Biosensing Techniques/methods , Nanotechnology/methods , Nanotubes, Carbon/chemistry , Animals , Biomarkers/chemistry , Biomarkers/metabolism , Biosensing Techniques/instrumentation , Cattle , Electric Conductivity , Limit of Detection , Models, Molecular , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Protein Conformation , Reproducibility of Results , Severe acute respiratory syndrome-related coronavirus , Streptavidin/metabolism , Transistors, Electronic
SELECTION OF CITATIONS
SEARCH DETAIL
...