Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Methods Mol Biol ; 2296: 381-392, 2021.
Article in English | MEDLINE | ID: mdl-33977460

ABSTRACT

Mass spectrometry is a sensitive and specific analytical technique that is capable of providing qualitative and quantitative data to resolve the protein elements of biochemical pathways that are altered by antibiotics. Here we present methods to study antibiotic susceptibility by changes in protein abundance, as exemplified by Klebsiella pneumoniae, a Gram-negative pathogen that colonizes mucosal surfaces of the human gastrointestinal and respiratory tracts. Cultured bacteria are exposed to antibiotics, the total proteomes of collected cell pellets are converted to complex peptide mixtures by filter-aided sample preparation (FASP), and the peptides are further processed by an optimized desalting procedure. A mixture of peptides from Klebsiella pneumoniae proteomes are analyzed by high-resolution mass spectrometry (MS) that is coupled to sensitive and comprehensive nano-liquid chromatography (nano-LC). The generic method described here for the identification and quantification of the proteome will provide a snapshot of differential protein abundances resulting from antimicrobial sensitivities, which can be used to model directed perturbations of the global system and to select targets of specific interest for further study.


Subject(s)
Anti-Bacterial Agents/pharmacology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Proteome/metabolism , Chromatography, Liquid/methods , Humans , Peptides/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods
2.
Antibodies (Basel) ; 9(3)2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32756297

ABSTRACT

Plague is a zoonotic disease that is caused by Yersinia pestis. Monoclonal antibodies (mAbs) that bind to the V-antigen, a virulence factor that is produced by Y. pestis, can passively protect mice from plague. An analysis of protective mAbs that bind to V-antigen was made to assess binding sites, avidities, and affinities. Anti-V mAbs were screened for their efficacy in a murine model of plague. Antigen-binding sites of protective V mAbs were determined with a linear peptide library, V-antigen fragment, competitive binding, and surface plasmon resonance. The avidities to the V-antigen was determined by ELISA, and affinities of the mAbs to the V-antigen were determined by surface plasmon resonance. The most protective mAb 7.3 bound to a unique conformational site on the V-antigen, while a less protective mAb bound to a different conformational site located on the same V-antigen fragment as mAb 7.3. The avidity of mAb 7.3 for the V-antigen was neither the strongest overall nor did it have the highest affinity for the V-antigen. The binding site of the most protective mAb was critical in its ability to protect against a lethal plague challenge.

3.
Mol Cell Proteomics ; 18(4): 657-668, 2019 04.
Article in English | MEDLINE | ID: mdl-30617156

ABSTRACT

Bacteria can circumvent the effect of antibiotics by transitioning to a poorly understood physiological state that does not involve conventional genetic elements of resistance. Here we examine antibiotic susceptibility with a Class A ß-lactamase+ invasive strain of Klebsiella pneumoniae that was isolated from a lethal outbreak within laboratory colonies of Chlorocebus aethiops sabaeus monkeys. Bacterial responses to the ribosomal synthesis inhibitors streptomycin and doxycycline resulted in distinct proteomic adjustments that facilitated decreased susceptibility to each antibiotic. Drug-specific changes to proteomes included proteins for receptor-mediated membrane transport and sugar utilization, central metabolism, and capsule production, whereas mechanisms common to both antibiotics included elevated scavenging of reactive oxygen species and turnover of misfolded proteins. Resistance to combined antibiotics presented integrated adjustments to protein levels as well as unique drug-specific proteomic features. Our results demonstrate that dampening of Klebsiella pneumoniae susceptibility involves global remodeling of the bacterial proteome to counter the effects of antibiotics and stabilize growth.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Klebsiella pneumoniae/metabolism , Proteome/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Cell Wall/drug effects , Cell Wall/metabolism , Chlorocebus aethiops , Drug Resistance, Microbial/drug effects , Intestine, Large/microbiology , Intestine, Large/pathology , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Microbial Sensitivity Tests , Proteomics , Ribosomes/drug effects , Ribosomes/metabolism
4.
ACS Infect Dis ; 4(12): 1705-1717, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30347144

ABSTRACT

Dengue is a mosquito-borne disease caused by four dengue virus serotypes (DENV1-4) that are loosely categorized by sequence commonalities and antibody recognition profiles. The highly variable envelope protein (E) that is prominently displayed on the surface of DENV is an essential component of vaccines currently under development, yet the impact of using single strains to represent each serotype in tetravalent vaccines has not been adequately studied. We synthesized chimeric E by replacing highly variable residues from a dengue virus serotype 2 vaccine strain (PUO-218) with those from 16 DENV2 lineages spanning 60 years of antigen evolution. Examining sera from human and rhesus macaques challenged with single strains of DENV2, antibody-E interactions were markedly inhibited or enhanced by residues mainly focused within a 480 Å2 footprint displayed on the E backbone. The striking impact of E diversity on polyclonal immune responses suggests that frequent antigen updates may be necessary for vaccines to counter shifts in circulating strains.


Subject(s)
Antibodies, Viral/immunology , Dengue Virus/immunology , Dengue/immunology , Adolescent , Adult , Animals , Antibodies, Neutralizing/immunology , Dengue/prevention & control , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Female , Humans , Macaca mulatta , Male , Middle Aged , Phylogeny , Serogroup , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Young Adult
5.
mSphere ; 3(2)2018.
Article in English | MEDLINE | ID: mdl-29577083

ABSTRACT

Mayaro virus (MAYV), Venezuelan equine encephalitis virus (VEEV), and chikungunya virus (CHIKV) are vector-borne alphaviruses that cocirculate in South America. Human infections by these viruses are frequently underdiagnosed or misdiagnosed, especially in areas with high dengue virus endemicity. Disease may progress to debilitating arthralgia (MAYV, CHIKV), encephalitis (VEEV), and death. Few standardized serological assays exist for specific human alphavirus infection detection, and antigen cross-reactivity can be problematic. Therefore, serological platforms that aid in the specific detection of multiple alphavirus infections will greatly expand disease surveillance for these emerging infections. In this study, serum samples from South American patients with PCR- and/or isolation-confirmed infections caused by MAYV, VEEV, and CHIKV were examined by using a protein microarray assembled with recombinant capsid, envelope protein 1 (E1), and E2 from nine New and Old World alphaviruses. Notably, specific antibody recognition of E1 was observed only with MAYV infections, whereas E2 was specifically targeted by antibodies from all of the alphavirus infections investigated, with evidence of cross-reactivity to E2 of o'nyong-nyong virus only in CHIKV-infected patient serum samples. Our findings suggest that alphavirus structural protein microarrays can distinguish infections caused by MAYV, VEEV, and CHIKV and that this multiplexed serological platform could be useful for high-throughput disease surveillance. IMPORTANCE Mayaro, chikungunya, and Venezuelan equine encephalitis viruses are closely related alphaviruses that are spread by mosquitos, causing diseases that produce similar influenza-like symptoms or more severe illnesses. Moreover, alphavirus infection symptoms can be similar to those of dengue or Zika disease, leading to underreporting of cases and potential misdiagnoses. New methods that can be used to detect antibody responses to multiple alphaviruses within the same assay would greatly aid disease surveillance efforts. However, possible antibody cross-reactivity between viruses can reduce the quality of laboratory results. Our results demonstrate that antibody responses to multiple alphaviruses can be specifically quantified within the same assay by using selected recombinant protein antigens and further show that Mayaro virus infections result in unique responses to viral envelope proteins.

6.
Clin Vaccine Immunol ; 24(4)2017 Apr.
Article in English | MEDLINE | ID: mdl-28228395

ABSTRACT

Zika virus (ZIKV) infections occur in areas where dengue virus (DENV), West Nile virus (WNV), yellow fever virus (YFV), and other viruses of the genus Flavivirus cocirculate. The envelope (E) proteins of these closely related flaviviruses induce specific long-term immunity, yet subsequent infections are associated with cross-reactive antibody responses that may enhance disease susceptibility and severity. To gain a better understanding of ZIKV infections against a background of similar viral diseases, we examined serological immune responses to ZIKV, WNV, DENV, and YFV infections of humans and nonhuman primates (NHPs). Using printed microarrays, we detected very specific antibody responses to primary infections with probes of recombinant E proteins from 15 species and lineages of flaviviruses pathogenic to humans, while high cross-reactivity between ZIKV and DENV was observed with 11 printed native viruses. Notably, antibodies from human primary ZIKV or secondary DENV infections that occurred in areas where flavivirus is endemic broadly recognized E proteins from many flaviviruses, especially DENV, indicating a strong influence of infection history on immune responses. A predictive algorithm was used to tentatively identify previous encounters with specific flaviviruses based on serum antibody interactions with the multispecies panel of E proteins. These results illustrate the potential impact of exposure to related viruses on the outcome of ZIKV infection and offer considerations for development of vaccines and diagnostics.


Subject(s)
Antibodies, Viral/blood , Antibody Formation , Cross Reactions , Endemic Diseases , Flaviviridae Infections/immunology , Flaviviridae Infections/veterinary , Animals , Humans , Macaca mulatta , Microarray Analysis , Primate Diseases/immunology , Protein Array Analysis
7.
Proteomics ; 17(9)2017 May.
Article in English | MEDLINE | ID: mdl-28198105

ABSTRACT

Extended spectrum beta-lactamase producing Klebsiella pneumoniae (ESBL-KP) causes life-threatening infections in susceptible and immuno-compromised individuals. Because of the emergence of multidrug resistance and tolerance, it is crucial to better understand the mechanisms by which ESBL-KP can adapt to antibiotic stress. The aim of this study was to provide an overview of the global proteome changes occurring in ESBL-KP in response to sub-lethal concentrations of the antibiotics doxycycline (DC, bacteriostatic) and streptomycin (SM, bactericidal), which both impair ribosomal synthesis of bacterial proteins. These results represent the greatest experimental coverage of the ESBL-KP proteome yet described. The 1538 proteins, representing 30% of the 5126 predicted KP gene products were identified from the combined experimental groups. Antibiotic stress resulted in significantly elevated levels of 42 proteins for DC and 55 for SM treatments, whereas 53 proteins were reduced for DC- and six for SM-treated bacteria. Specifically, the ESBL-KP response to DC was accompanied by the reduced levels of the porins LamB, CirA, FepA, and OmpC. In contrast to DC, the stress response to SM demonstrated a dramatic increase in the peroxidase detoxification pathway proteins PutA, KatG, KatE, and Dps, which prevent harmful hydroxyl radical formation. The results from this proteomic study are important for understanding adaptive responses to antibiotics, and may provide novel targets for the development of new therapeutic strategies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/analysis , Gene Expression Regulation, Bacterial/drug effects , Klebsiella pneumoniae/metabolism , Proteome/analysis , Proteomics/methods , beta-Lactamases/metabolism , Bacterial Proteins/metabolism , Doxycycline/pharmacology , Humans , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Streptomycin/pharmacology
8.
Mol Cell Proteomics ; 15(10): 3220-3232, 2016 10.
Article in English | MEDLINE | ID: mdl-27489291

ABSTRACT

Binary protein interactions form the basic building blocks of molecular networks and dynamic assemblies that control all cellular functions of bacteria. Although these protein interactions are a potential source of targets for the development of new antibiotics, few high-confidence data sets are available for the large proteomes of most pathogenic bacteria. We used a library of recombinant proteins from the plague bacterium Yersinia pestis to probe planar microarrays of immobilized proteins that represented ∼85% (3552 proteins) of the bacterial proteome, resulting in >77,000 experimentally determined binary interactions. Moderate (KD ∼µm) to high-affinity (KD ∼nm) interactions were characterized for >1600 binary complexes by surface plasmon resonance imaging of microarrayed proteins. Core binary interactions that were in common with other gram-negative bacteria were identified from the results of both microarray methods. Clustering of proteins within the interaction network by function revealed statistically enriched complexes and pathways involved in replication, biosynthesis, virulence, metabolism, and other diverse biological processes. The interaction pathways included many proteins with no previously known function. Further, a large assembly of proteins linked to transcription and translation were contained within highly interconnected subregions of the network. The two-tiered microarray approach used here is an innovative method for detecting binary interactions, and the resulting data will serve as a critical resource for the analysis of protein interaction networks that function within an important human pathogen.


Subject(s)
Bacterial Proteins/metabolism , Protein Array Analysis/methods , Yersinia pestis/metabolism , Cell-Free System , Cluster Analysis , Computational Biology/methods , Protein Interaction Maps , Proteomics/methods , Surface Plasmon Resonance
9.
Clin Vaccine Immunol ; 23(8): 717-24, 2016 08.
Article in English | MEDLINE | ID: mdl-27335383

ABSTRACT

A detailed understanding of serological immune responses to Ebola and Marburg virus infections will facilitate the development of effective diagnostic methods, therapeutics, and vaccines. We examined antibodies from Ebola or Marburg survivors 1 to 14 years after recovery from disease, by using a microarray that displayed recombinant nucleoprotein (NP), viral protein 40 (VP40), envelope glycoprotein (GP), and inactivated whole virions from six species of filoviruses. All three outbreak cohorts exhibited significant antibody responses to antigens from the original infecting species and a pattern of additional filoviruses that varied by outbreak. NP was the most cross-reactive antigen, while GP was the most specific. Antibodies from survivors of infections by Marburg marburgvirus (MARV) species were least cross-reactive, while those from survivors of infections by Sudan virus (SUDV) species exhibited the highest cross-reactivity. Based on results revealed by the protein microarray, persistent levels of antibodies to GP, NP, and VP40 were maintained for up to 14 years after infection, and survival of infection caused by one species imparted cross-reactive antibody responses to other filoviruses.


Subject(s)
Antibodies, Viral/blood , Cross Reactions , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Marburg Virus Disease/immunology , Marburgvirus/immunology , Animals , Antibody Formation , Antigens, Viral/immunology , Cohort Studies , Disease Outbreaks , Healthy Volunteers , Hemorrhagic Fever, Ebola/epidemiology , Humans , Marburg Virus Disease/epidemiology , Microarray Analysis , Protein Array Analysis , Survivors , Viral Structural Proteins/genetics
10.
PLoS One ; 10(8): e0134984, 2015.
Article in English | MEDLINE | ID: mdl-26302245

ABSTRACT

Protein tyrosine phosphatases dephosphorylate tyrosine residues of proteins, whereas, dual specificity phosphatases (DUSPs) are a subgroup of protein tyrosine phosphatases that dephosphorylate not only Tyr(P) residue, but also the Ser(P) and Thr(P) residues of proteins. The DUSPs are linked to the regulation of many cellular functions and signaling pathways. Though many cellular targets of DUSPs are known, the relationship between catalytic activity and substrate specificity is poorly defined. We investigated the interactions of peptide substrates with select DUSPs of four types: MAP kinases (DUSP1 and DUSP7), atypical (DUSP3, DUSP14, DUSP22 and DUSP27), viral (variola VH1), and Cdc25 (A-C). Phosphatase recognition sites were experimentally determined by measuring dephosphorylation of 6,218 microarrayed Tyr(P) peptides representing confirmed and theoretical phosphorylation motifs from the cellular proteome. A broad continuum of dephosphorylation was observed across the microarrayed peptide substrates for all phosphatases, suggesting a complex relationship between substrate sequence recognition and optimal activity. Further analysis of peptide dephosphorylation by hierarchical clustering indicated that DUSPs could be organized by substrate sequence motifs, and peptide-specificities by phylogenetic relationships among the catalytic domains. The most highly dephosphorylated peptides represented proteins from 29 cell-signaling pathways, greatly expanding the list of potential targets of DUSPs. These newly identified DUSP substrates will be important for examining structure-activity relationships with physiologically relevant targets.


Subject(s)
Dual-Specificity Phosphatases/metabolism , Phosphotyrosine/metabolism , Amino Acid Motifs , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/metabolism , Dual Specificity Phosphatase 3/genetics , Dual Specificity Phosphatase 3/metabolism , Dual-Specificity Phosphatases/genetics , Humans , Mitogen-Activated Protein Kinase Phosphatases/genetics , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phylogeny , Protein Array Analysis , Recombinant Proteins , Signal Transduction , Substrate Specificity , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
11.
Mol Cell Proteomics ; 8(5): 924-35, 2009 May.
Article in English | MEDLINE | ID: mdl-19112181

ABSTRACT

Antibodies provide a sensitive indicator of proteins displayed by bacteria during sepsis. Because signals produced by infection are naturally amplified during the antibody response, host immunity can be used to identify biomarkers for proteins that are present at levels currently below detectable limits. We developed a microarray comprising approximately 70% of the 4066 proteins contained within the Yersinia pestis proteome to identify antibody biomarkers distinguishing plague from infections caused by other bacterial pathogens that may initially present similar clinical symptoms. We first examined rabbit antibodies produced against proteomes extracted from Y. pestis, Burkholderia mallei, Burkholderia cepecia, Burkholderia pseudomallei, Pseudomonas aeruginosa, Salmonella typhimurium, Shigella flexneri, and Escherichia coli, all pathogenic Gram-negative bacteria. These antibodies enabled detection of shared cross-reactive proteins, fingerprint proteins common for two or more bacteria, and signature proteins specific to each pathogen. Recognition by rabbit and non-human primate antibodies involved less than 100 of the thousands of proteins present within the Y. pestis proteome. Further antigen binding patterns were revealed that could distinguish plague from anthrax, caused by the Gram-positive bacterium Bacillus anthracis, using sera from acutely infected or convalescent primates. Thus, our results demonstrate potential biomarkers that are either specific to one strain or common to several species of pathogenic bacteria.


Subject(s)
Antibodies, Bacterial/immunology , Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/immunology , Protein Array Analysis , Proteome/analysis , Animals , Antibodies, Bacterial/metabolism , Antibody Formation/immunology , Bacterial Proteins/metabolism , Cross Reactions/immunology , Macaca mulatta/immunology , Macaca mulatta/microbiology , Plague/immunology , Protein Binding , Proteome/immunology , Rabbits , Yersinia pestis/immunology
12.
Proteomics Clin Appl ; 2(10-11): 1528-38, 2008 Oct.
Article in English | MEDLINE | ID: mdl-21136800

ABSTRACT

Control of smallpox by mass vaccination was one of the most effective public health measures ever employed for eradicating a devastating infectious disease. However, new methods are needed for monitoring smallpox immunity within current vulnerable populations, and for the development of replacement vaccines for use by immunocompromized or low-responding individuals. As a measure for achieving this goal, we developed a protein microarray of the vaccinia virus proteome by using high-throughput baculovirus expression and purification of individual elements. The array was validated with therapeutic-grade, human hyperimmune sera, and these data were compared to results obtained from individuals vaccinated against smallpox using Dryvax. A high level of reproducibility with a very low background were apparent in repetitive assays that confirmed previously reported antigens and identified new proteins that may be important for neutralizing viral infection. Our results suggest that proteins recognized by antibodies from all vaccinees constituted <10% of the total vaccinia proteome.

SELECTION OF CITATIONS
SEARCH DETAIL
...