Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 41(21): 3308-3316, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37085457

RESUMO

In the past decade, camelid nanobodies have been developed for multiple applications, including immuno-imaging, cancer immunotherapy, and antiviral therapeutics. Despite the prevalence of these approaches, nanobodies have rarely been used to assess the potency of vaccine antigen candidates, which are primarily based on mAb binding approaches. In this work, we demonstrate that a nanobody-based ELISA method is suitable for characterization of a leading respiratory syncytial virus (RSV) vaccine candidate, RSVPreF3. This nanobody, F-VHH-L66, compares similarly with AM14, an antibody well-known to be specific for the prefusion form of the RSV surface fusion glycoprotein, RSV F. ELISA assays based on F-VHH-L66 were specific for the trimeric, prefusion form of RSV F, the antigen conformation that best generates neutralizing antibodies. Additionally, the F-VHH-L66-based ELISA proved accurate, linear, and stability-indicating. Statistical analysis of 65 independent F-VHH-L66-based ELISA experiments indicated assay performance similar to that of ELISA assays based on AM14. Moreover, the binding kinetics of F-VHH-L66 to RSVPreF3 are comparable to those of AM14 when measured by surface plasmon resonance (SPR). Finally, F-VHH-L66 neutralized RSV(A) with similar efficacy as AM14; this bioactivity data further supports its use as an alternative to AM14 for pre-fusion specific structural characterization of RSVPreF3.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Vírus Sincicial Respiratório Humano , Anticorpos de Domínio Único , Humanos , Anticorpos Antivirais , Anticorpos Neutralizantes , Antivirais , Proteínas Virais de Fusão , Infecções por Vírus Respiratório Sincicial/prevenção & controle
2.
Biotechnol Bioeng ; 114(8): 1753-1761, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28369693

RESUMO

Human neurturin (NTN) is a cystine knot growth factor with potential therapeutic use in diseases such as Parkinson's and diabetes. Scalable high titer production of native NTN is particularly challenging because of the cystine knot structure which consists of an embedded ring comprised of at least three disulfide bonds. We sought to pursue enhanced scalable production of NTN in Escherichia coli. Our initial efforts focused on codon optimization of the first two codons following AUG, but these studies resulted in only a marginal increase in NTN expression. Therefore, we pursued an alternative strategy of using a bicistronic vector for NTN expression designed to reduce mRNA secondary structure to achieve increased ribosome binding and re-initiation. The first cistron was designed to prevent sequestration of the translation initiation region in a secondary conformation. The second cistron, which contained the NTN coding sequence itself, was engineered to disrupt double bonded base pairs and destabilize the secondary structure for ribosome re-initiation. The ensemble approach of reducing NTN's mRNA secondary structure and using the bicistronic vector had an additive effect resulting in significantly increased NTN expression. Our strain selection studies were conducted in a miniaturized bioreactor. An optimized strain was selected and scaled up to a 100 L fermentor, which yielded an inclusion body titer of 2 g/L. The inclusion bodies were refolded to yield active NTN. We believe that our strategy is applicable to other candidate proteins that are difficult-to-express due to stable mRNA secondary structures. Biotechnol. Bioeng. 2017;114: 1753-1761. © 2017 Wiley Periodicals, Inc.


Assuntos
Escherichia coli/fisiologia , Éxons/genética , Melhoramento Genético/métodos , Vetores Genéticos/genética , Neurturina/biossíntese , RNA Mensageiro/genética , Regulação Bacteriana da Expressão Gênica/genética , Genes/genética , Neurturina/genética , Relação Estrutura-Atividade , Regulação para Cima/genética
3.
ACS Synth Biol ; 6(7): 1370-1379, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28350472

RESUMO

Chinese Hamster Ovary (CHO) cells are routinely optimized to stably express monoclonal antibodies (mAbs) at high titers. At the early stages of lead isolation and optimization, hundreds of sequences for the target protein of interest are screened. Typically, cell-based transient expression technology platforms are used for expression screening, but these can be time- and resource-intensive. Here, we have developed a cell-free protein synthesis (CFPS) platform utilizing a commercially available CHO extract for the rapid in vitro synthesis of active, aglycosylated mAbs. Specifically, we optimized reaction conditions to maximize protein yields, established an oxidizing environment to enable disulfide bond formation, and demonstrated the importance of temporal addition of heavy chain and light chain plasmids for intact mAb production. Using our optimized platform, we demonstrate for the first time to our knowledge the cell-free synthesis of biologically active, intact mAb at >100 mg/L using a eukaryotic-based extract. We then explored the utility of our system as a tool for ranking yields of candidate antibodies. Unlike stable or transient transfection-based screening, which requires a minimum of 7 days for setup and execution, results using our CHO-based CFPS platform are attained within 2 days and it is well-suited for automation. Further development would provide a tool for rapid, high-throughput prediction of mAb expression ranking to accelerate design-build-test cycles required for antibody expression and engineering. Looking forward, the CHO-based CFPS platform could facilitate the synthesis of toxic proteins as well.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/metabolismo , Animais , Biotecnologia/métodos , Células CHO , Sistema Livre de Células , Cricetulus , Biossíntese de Proteínas
4.
PLoS One ; 11(12): e0167935, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28002433

RESUMO

Humans and higher primates are unique in that they lack uricase, the enzyme capable of oxidizing uric acid. As a consequence of this enzyme deficiency, humans have high serum uric acid levels. In some people, uric acid levels rise above the solubility limit resulting in crystallization in joints, acute inflammation in response to those crystals causes severe pain; a condition known as gout. Treatment for severe gout includes injection of non-human uricase to reduce serum uric acid levels. Krystexxa® is a hyper-PEGylated pig-baboon chimeric uricase indicated for chronic refractory gout that induces an immunogenic response in 91% of treated patients, including infusion reactions (26%) and anaphylaxis (6.5%). These properties limit its use and effectiveness. An innovative approach has been used to develop a therapeutic uricase with improved properties such as: soluble expression, neutral pH solubility, high E. coli expression level, thermal stability, and excellent activity. More than 200 diverse uricase sequences were aligned to guide protein engineering and reduce putative sequence liabilities. A single uricase lead candidate was identified, which showed low potential for immunogenicity in >200 human donor samples selected to represent diverse HLA haplotypes. Cysteines were engineered into the lead sequence for site specific PEGylation and studies demonstrated >95% PEGylation efficiency. PEGylated uricase retains enzymatic activity in vitro at neutral pH, in human serum and in vivo (rats and canines) and has an extended half-life. In canines, an 85% reduction in serum uric acid levels was observed with a single subcutaneous injection. This PEGylated, non-immunogenic uricase has the potential to provide meaningful benefits to patients with gout.


Assuntos
Gota/tratamento farmacológico , Urato Oxidase/uso terapêutico , Animais , Varredura Diferencial de Calorimetria , Cães , Escherichia coli/metabolismo , Meia-Vida , Humanos , Concentração de Íons de Hidrogênio , Cinética , Papio , Polietilenoglicóis/química , Ratos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico , Especificidade por Substrato , Suínos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Urato Oxidase/efeitos adversos , Urato Oxidase/imunologia
5.
Appl Microbiol Biotechnol ; 97(6): 2627-38, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23053069

RESUMO

Quorum sensing (QS), the process of autoinducer-mediated cell-cell signaling among bacteria, facilitates biofilm formation, virulence, and many other multicellular phenotypes. QS inhibitors are being investigated as antimicrobials because of their potential to reduce symptoms of infectious disease while slowing the emergence of resistant strains. Autoinducer-2 (AI-2) analogs have been shown to inhibit genotypic QS responses among many bacteria. We demonstrate for the first time, the ability of C1-alkyl AI-2 analog, isobutyl-DPD, to significantly inhibit the maturation of Escherichia coli biofilms grown in vitro. Using a novel microfluidic device that incorporates dynamic, real-time measurements of biofilm density, we also show that a combinatorial approach wherein isobutyl-DPD ((S)-4,5-dihydroxy-2,3-pentanedione) is used with the antibiotic gentamicin is quite effective in rendering near complete clearance of pre-existing E. coli biofilms. Similarly, another AI-2 analog, phenyl-DPD, also used in combination with near MIC levels of gentamicin, resulted in clearance of preformed Pseudomonas aeruginosa biofilms. Clearance of pre-existing biofilms has remained a significant health care challenge; these results warrant consideration of a new approach based on the combination of "quenching" QS signal transduction processes with traditional antibiotic treatment.


Assuntos
Antibacterianos/metabolismo , Biofilmes/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/fisiologia , Homosserina/análogos & derivados , Lactonas/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , Biofilmes/crescimento & desenvolvimento , Escherichia coli/crescimento & desenvolvimento , Gentamicinas/metabolismo , Homosserina/metabolismo , Pseudomonas aeruginosa/crescimento & desenvolvimento
6.
ACS Chem Biol ; 7(6): 1023-30, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22433054

RESUMO

There have been intensive efforts to find small molecule antagonists for bacterial quorum sensing (QS) mediated by the "universal" QS autoinducer, AI-2. Previous work has shown that linear and branched acyl analogues of AI-2 can selectively modulate AI-2 signaling in bacteria. Additionally, LsrK-dependent phosphorylated analogues have been implicated as the active inhibitory form against AI-2 signaling. We used these observations to synthesize an expanded and diverse array of AI-2 analogues, which included aromatic as well as cyclic C-1-alkyl analogues. Species-specific analogues that disrupted AI-2 signaling in Escherichia coli and Salmonella typhimurium were identified. Similarly, analogues that disrupted QS behaviors in Pseudomonas aeruginosa were found. Moreover, we observed a strong correlation between LsrK-dependent phosphorylation of these acyl analogues and their ability to suppress QS. Significantly, we demonstrate that these analogues can selectively antagonize QS in single bacterial strains in a physiologically relevant polymicrobial culture.


Assuntos
Escherichia coli/metabolismo , Homosserina/análogos & derivados , Lactonas/química , Lactonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Percepção de Quorum , Salmonella typhimurium/metabolismo , Escherichia coli/enzimologia , Proteínas de Escherichia coli/metabolismo , Homosserina/síntese química , Homosserina/química , Homosserina/metabolismo , Lactonas/síntese química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Repressoras/metabolismo , Salmonella typhimurium/enzimologia , Transdução de Sinais , beta-Galactosidase/metabolismo
7.
Enzyme Microb Technol ; 49(2): 113-23, 2011 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22112397

RESUMO

Bacteria have been evolving antibiotic resistance since their discovery in the early twentieth century. Most new antibiotics are derivatives of older generations and there are now bacteria that are virtually resistant to almost all antibiotics. This poses a global threat to human health and has been classified as a clinical "super-challenge", which has necessitated research into new antimicrobials that inhibit bacterial virulence while minimizing selective pressures that lead to the emergence of resistant strains. Quorum sensing (QS), the process of population dependent bacterial cell-cell signaling, can accelerate bacterial virulence and is an increasingly interesting target for developing next generation antimicrobials. Most QS inhibitors target species-specific signals, such as acylhomoserine lactones (AHLs) and oligopeptides. Methodologies for intercepting the cross-species signal, autoinducer-2 (AI-2), have only recently emerged. We review these strategies to prevent the relay of the AI-2 signal amongst pathogens, including Escherichia coli, Salmonella enterica serovar Typhimurium, Vibrio cholerae and Pseudomonas aeruginosa. Inhibition mechanisms are categorized based on the target (i.e., enzymes for signal generation, the signal molecule itself, or the various components of the signal transduction process). The universal nature of the AI-2 signal imparts on its inhibitors the potential for broad spectrum use.


Assuntos
Anti-Infecciosos/farmacologia , Homosserina/análogos & derivados , Lactonas/antagonistas & inibidores , Percepção de Quorum/efeitos dos fármacos , Anti-Infecciosos/química , Bactérias/efeitos dos fármacos , Bactérias/patogenicidade , Fenômenos Fisiológicos Bacterianos/efeitos dos fármacos , Descoberta de Drogas , Homosserina/antagonistas & inibidores , Homosserina/fisiologia , Humanos , Percepção de Quorum/fisiologia
8.
Biochemistry ; 50(32): 6983-93, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21736305

RESUMO

Quorum sensing is an intricate example of "social" behavior in microbial communities mediated by small secreted molecules (autoinducers). The mechanisms of membrane permeation remain elusive for many of them. Here we present the assessment of membrane permeability for three natural autoinducers and four synthetic analogues based on their polarity, surface activity, affinity for lipid monolayers, and ability to induce lateral pressure changes in the inner E. coli membrane sensed by the bacterial tension-activated channel MscS. AI-1 (N-(3-oxodecanoyl)-l-homoserine lactone) is surface-active, and it robustly inserts into lipid monolayers, indicating strong propensity toward membranes. When presented to membrane patches from the cytoplasmic side, AI-1 transiently shifts MscS's activation curve toward higher tensions due to intercalation into the cytoplasmic leaflet followed by redistribution to the opposite side. Indole showed no detectable surface activity at the air-water interface but produced a moderate increase of lateral pressure in monolayers and was potent at shifting activation curves of MscS, demonstrating transients on sequential additions. AI-2 (4,5-dihydroxy-2,3-pentanedione, DPD) showed little activity at the interfaces, correspondingly with no effect on MscS activation. After chemical modification with isobutyl, hexyl, or heptyl chains, AI-2 displayed strong surface activity. Hexyl and especially heptyl AI-2 induced robust transient shifts of MscS activation curves. The data strongly suggest that both AI-1 and indole are directly permeable through the membrane. AI-2, more hydrophilic, shows low affinity toward lipids and thus requires a transport system, whereas alkyl analogues of AI-2 should permeate the membrane directly.


Assuntos
Escherichia coli/fisiologia , Percepção de Quorum , Permeabilidade da Membrana Celular , Tensão Superficial
9.
Anal Chem ; 83(15): 5920-7, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21688780

RESUMO

We present a unique microfluidic platform to allow for quick and sensitive probing of protein adsorption to various functionalized surfaces. The ability to tailor a sensor surface for a specific analyte is crucial for the successful application of portable gas and fluid sensors and is of great interest to the drug screening community. However, choosing the correct surface chemistry to successfully passivate against nonspecific binding typically requires repeated trial and error experiments. The presented device incorporates an array of integrated electrochemical sensors for fast, sensitive, label-free detection of these binding interactions. The layout of the electrodes allows for loading various surface chemistries in one direction while sensing their interactions with particular compounds in another without any cross-contamination. Impedance data is collected for three commonly used passivation compounds (mercaptohexanol, polyethylene glycol, and bovine serum albumin) and demonstrates their interaction with three commonly studied proteins in genetic and cancer research (cAMP receptor protein, tumor necrosis factor α, and tumor necrosis factor ß). The ability to quickly characterize various surface interactions provides knowledge for selecting optimal functionalization for any biosensor.


Assuntos
Proteína Receptora de AMP Cíclico/metabolismo , Técnicas Eletroquímicas/métodos , Linfotoxina-alfa/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Soroalbumina Bovina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Técnicas Biossensoriais/métodos , Bovinos , Proteína Receptora de AMP Cíclico/química , Eletrodos , Linfotoxina-alfa/química , Técnicas Analíticas Microfluídicas/instrumentação , Polietilenoglicóis/química , Mapeamento de Interação de Proteínas , Soroalbumina Bovina/química , Propriedades de Superfície , Fator de Necrose Tumoral alfa/química
10.
J Am Chem Soc ; 132(32): 11141-50, 2010 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-20698680

RESUMO

The widespread use of antibiotics and the emergence of resistant strains call for new approaches to treat bacterial infection. Bacterial cell-cell communication or "quorum sensing" (QS) is mediated by "signatures" of small molecules that represent targets for "quenching" communication and avoiding virulent phenotypes. Only a handful of small molecules that antagonize the action of the "universal" autoinducer, AI-2, have been reported. The biological basis of antagonism, as well as the targets for these select few AI-2 antagonists, have not been clearly defined. We have developed C-1 alkyl analogs of AI-2 that quench the QS response in multiple bacterial species simultaneously. We also demonstrate the biological basis for this action. Like AI-2, the analogs are activated by the bacterial kinase, LsrK, and modulate AI-2 specific gene transcription through the transcriptional regulator, LsrR. Interestingly, addition of a single carbon to the C1-alkyl chain of the analog plays a crucial role in determining the effect of the analog on the QS response. While an ethyl modified analog is an agonist, propyl becomes an antagonist of the QS circuit. In a trispecies synthetic ecosystem comprised of E. coli, S. typhimurium, and V. harveyi we discovered both cross-species and species-specific anti-AI-2 QS activities. Our results suggest entirely new modalities for interrupting or tailoring the network of communication among bacteria.


Assuntos
Bactérias/citologia , Bactérias/efeitos dos fármacos , Homosserina/análogos & derivados , Lactonas/química , Lactonas/metabolismo , Pentanos/síntese química , Pentanos/farmacologia , Percepção de Quorum/efeitos dos fármacos , Bactérias/metabolismo , Transporte Biológico , Ecossistema , Homosserina/antagonistas & inibidores , Homosserina/química , Homosserina/metabolismo , Lactonas/antagonistas & inibidores , Modelos Moleculares , Conformação Molecular , Pentanos/química , Pentanos/metabolismo , Fosforilação
11.
Nat Nanotechnol ; 5(3): 213-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20081846

RESUMO

Biological nanofactories, which are engineered to contain modules that can target, sense and synthesize molecules, can trigger communication between different bacterial populations. These communications influence biofilm formation, virulence, bioluminescence and many other bacterial functions in a process called quorum sensing. Here, we show the assembly of a nanofactory that can trigger a bacterial quorum sensing response in the absence of native quorum molecules. The nanofactory comprises an antibody (for targeting) and a fusion protein that produces quorum molecules when bound to the targeted bacterium. Our nanofactory selectively targets the appropriate bacteria and triggers a quorum sensing response when added to two populations of bacteria. The nanofactories also trigger communication between two bacterial populations that are otherwise non-communicating. We envision the use of these nanofactories in generating new antimicrobial treatments that target the communication networks of bacteria rather than their viability.


Assuntos
Anticorpos Antibacterianos/farmacologia , Homosserina/análogos & derivados , Lactonas/farmacologia , Nanoestruturas/química , Nanotecnologia/métodos , Percepção de Quorum/efeitos dos fármacos , Anticorpos Antibacterianos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Escherichia coli/metabolismo , Citometria de Fluxo , Homosserina/metabolismo , Homosserina/farmacologia , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Lactonas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Salmonella/metabolismo
12.
ACS Chem Biol ; 5(2): 223-32, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20025244

RESUMO

Bacterial quorum sensing (QS) is a cell-cell communication process, mediated by signaling molecules, that alters various phenotypes including pathogenicity. Methods to interrupt these communication networks are being pursued as next generation antimicrobials. We present a technique for interrupting communication among bacteria that exploits their native and highly specific machinery for processing the signaling molecules themselves. Specifically, our approach is to bring native intracellular signal processing mechanisms to the extracellular surroundings and "quench" crosstalk among a variety of strains. In this study, the QS system based on the interspecies signaling molecule autoinducer-2 (AI-2) is targeted because of its prevalence among prokaryotes (it functions in over 80 bacterial species). We demonstrate that the Escherichia coli AI-2 kinase, LsrK, can phosphorylate AI-2 in vitro, and when LsrK-treated AI-2 is added ex vivo to E. coli populations, the native QS response is significantly reduced. Further, LsrK-mediated degradation of AI-2 attenuates the QS response among Salmonella typhimurium and Vibrio harveyi even though the AI-2 signal transduction mechanisms and the phenotypic responses are species-specific. Analogous results are obtained from a synthetic ecosystem where three species of bacteria (enteric and marine) are co-cultured. Finally, the addition of LsrK and ATP to growing co-cultures of E. coli and S. typhimurium exhibits significantly reduced native "cross-talk" that ordinarily exists among and between species in an ecosystem. We believe this nature-inspired enzymatic approach for quenching QS systems will spawn new methods for controlling cell phenotype and potentially open new avenues for controlling bacterial pathogenicity.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Homosserina/análogos & derivados , Lactonas/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Percepção de Quorum , Técnicas de Cocultura , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Proteínas de Escherichia coli/farmacologia , Homosserina/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/enzimologia , Salmonella typhimurium/crescimento & desenvolvimento , Vibrio/efeitos dos fármacos , Vibrio/enzimologia , Vibrio/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA