Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Am J Trop Med Hyg ; 92(1): 9-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25294612

ABSTRACT

Parasite antigen diversity poses an obstacle to developing an effective malaria vaccine. A protein microarray containing Plasmodium falciparum apical membrane antigen 1 (AMA1, n = 57) and merozoite surface protein 1 19-kD (MSP119, n = 10) variants prevalent at a malaria vaccine testing site in Bandiagara, Mali, was used to assess changes in seroreactivity caused by seasonal and lifetime exposure to malaria. Malian adults had significantly higher magnitude and breadth of seroreactivity to variants of both antigens than did Malian children. Seroreactivity increased over the course of the malaria season in children and adults, but the difference was more dramatic in children. These results help to validate diversity-covering protein microarrays as a promising tool for measuring the breadth of antibody responses to highly variant proteins, and demonstrate the potential of this new tool to help guide the development of malaria vaccines with strain-transcending efficacy.


Subject(s)
Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Seasons , Adult , Animals , Child , Humans
2.
Sci Rep ; 3: 3549, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24351974

ABSTRACT

Complete sterile protection to Plasmodium falciparum (Pf) infection mediated by pre-erythrocytic immunity can be experimentally induced under chloroquine prophylaxis, through immunization with sporozoites from infected mosquitoes' bites (CPS protocol). To characterize the profile of CPS induced antibody (Ab) responses, we developed a proteome microarray containing 809 Pf antigens showing a distinct Ab profile with recognition of antigens expressed in pre-erythrocytic life-cycle stages. In contrast, plasma from naturally exposed semi-immune individuals from Kenya was skewed toward antibody reactivity against asexual blood stage antigens. CPS-immunized and semi-immune individuals generated antibodies against 192 and 202 Pf antigens, respectively, but only 60 antigens overlapped between the two groups. Although the number of reactive antigens varied between the CPS-immunized individuals, all volunteers reacted strongly against the pre-erythrocytic antigens circumsporozoite protein (CSP) and liver stage antigen 1 (LSA1). Well classified merozoite and erythrocytic antigens were strongly reactive in semi-immune individuals but lacking in the CPS immunized group. These data show that the antibody profile of CPS-immunized and semi-immune groups have quite distinct profiles reflecting their protective immunity; antibodies from CPS immunized individuals react strongly against pre-erythrocytic while semi-immune individuals mainly react against erythrocytic antigens.


Subject(s)
Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Antibodies/immunology , Antibody Formation/immunology , Antimalarials/therapeutic use , Chloroquine/therapeutic use , Erythrocytes/parasitology , Healthy Volunteers , Humans , Kenya , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Protein Array Analysis
3.
J Infect Dis ; 208(9): 1514-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23901079

ABSTRACT

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) antigens mediate parasite sequestration and host immune evasion. Reactivity to 21 PfEMP1 fragments on a protein microarray was measured in serum samples from Malian children aged 1-6 years and adults. Seroreactivity to PfEMP1 fragments was higher in adults than in children; intracellular conserved fragments were more widely recognized than were extracellular hypervariable fragments. Over a malaria season, children maintained this differential seroreactivity and recognized additional intracellular PfEMP1 fragments. This approach has the potential to identify conserved, seroreactive extracellular PfEMP1 domains critical for protective immunity to malaria.


Subject(s)
Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Peptide Fragments/immunology , Protozoan Proteins/immunology , Adult , Antibodies, Protozoan/blood , Case-Control Studies , Child , Child, Preschool , Humans , Infant , Malaria, Falciparum/blood , Plasmodium falciparum/immunology , Protein Array Analysis , Protein Structure, Tertiary
4.
PLoS One ; 8(3): e57587, 2013.
Article in English | MEDLINE | ID: mdl-23505434

ABSTRACT

Pemphigus vulgaris (PV) is a mucocutaneous blistering disease characterized by IgG autoantibodies against the stratified squamous epithelium. Current understanding of PV pathophysiology does not explain the mechanism of acantholysis in patients lacking desmoglein antibodies, which justifies a search for novel targets of pemphigus autoimmunity. We tested 264 pemphigus and 138 normal control sera on the multiplexed protein array platform containing 701 human genes encompassing many known keratinocyte cell-surface molecules and members of protein families targeted by organ-non-specific PV antibodies. The top 10 antigens recognized by the majority of test patients' sera were proteins encoded by the DSC1, DSC3, ATP2C1, PKP3, CHRM3, COL21A1, ANXA8L1, CD88 and CHRNE genes. The most common combinations of target antigens included at least one of the adhesion molecules DSC1, DSC3 or PKP3 and/or the acetylcholine receptor CHRM3 or CHRNE with or without the MHC class II antigen DRA. To identify the PV antibodies most specific to the disease process, we sorted the data based on the ratio of patient to control frequencies of antigen recognition. The frequency of antigen recognition by patients that exceeded that of control by 10 and more times were the molecules encoded by the CD33, GP1BA, CHRND, SLC36A4, CD1B, CD32, CDH8, CDH9, PMP22 and HLA-E genes as well as mitochondrial proteins encoded by the NDUFS1, CYB5B, SOD2, PDHA1 and FH genes. The highest specificity to PV showed combinations of autoantibodies to the calcium pump encoded by ATP2C1 with C5a receptor plus DSC1 or DSC3 or HLA-DRA. The results identified new targets of pemphigus autoimmunity. Novel autoantibody signatures may help explain individual variations in disease severity and treatment response, and serve as sensitive and specific biomarkers for new diagnostic assays in PV patients.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Pemphigus/blood , Pemphigus/immunology , Proteomics , Antibody Specificity , Autoantigens/immunology , Desmoglein 3/immunology , Humans , Principal Component Analysis , Protein Array Analysis , Sensitivity and Specificity
5.
Sci Rep ; 3: 1043, 2013.
Article in English | MEDLINE | ID: mdl-23304434

ABSTRACT

Current serological diagnostic assays for typhoid fever are based on detecting antibodies against Salmonella LPS or flagellum, resulting in a high false-positive rate. Here we used a protein microarray containing 2,724 Salmonella enterica serovar Typhi antigens (>63% of proteome) and identified antibodies against 16 IgG antigens and 77 IgM antigens that were differentially reactive among acute typhoid patients and healthy controls. The IgG target antigens produced a sensitivity of 97% and specificity of 80%, whereas the IgM target antigens produced 97% and 91% sensitivity and specificity, respectively. Our analyses indicated certain features such as membrane association, secretion, and protein expression were significant enriching features of the reactive antigens. About 72% of the serodiagnostic antigens were within the top 25% of the ranked antigen list using a Naïve bayes classifier. These data provide an important resource for improved diagnostics, therapeutics and vaccine development against an important human pathogen.


Subject(s)
Antibodies, Bacterial/analysis , Antigens/immunology , Protein Array Analysis , Salmonella typhi/metabolism , Typhoid Fever/diagnosis , Antigens/genetics , Antigens/metabolism , Biomarkers/analysis , Cloning, Molecular , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Proteome/immunology , Proteome/metabolism , ROC Curve , Sensitivity and Specificity
6.
J Virol ; 86(8): 4328-39, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22318154

ABSTRACT

Routine serodiagnosis of herpes simplex virus (HSV) infections is currently performed using recombinant glycoprotein G (gG) antigens from herpes simplex virus 1 (HSV-1) and HSV-2. This is a single-antigen test and has only one diagnostic application. Relatively little is known about HSV antigenicity at the proteome-wide level, and the full potential of mining the antibody repertoire to identify antigens with other useful diagnostic properties and candidate vaccine antigens is yet to be realized. To this end we produced HSV-1 and -2 proteome microarrays in Escherichia coli and probed them against a panel of sera from patients serotyped using commercial gG-1 and gG-2 (gGs for HSV-1 and -2, respectively) enzyme-linked immunosorbent assays. We identified many reactive antigens in both HSV-1 and -2, some of which were type specific (i.e., recognized by HSV-1- or HSV-2-positive donors only) and others of which were nonspecific or cross-reactive (i.e., recognized by both HSV-1- and HSV-2-positive donors). Both membrane and nonmembrane virion proteins were antigenic, although type-specific antigens were enriched for membrane proteins, despite being expressed in E. coli.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/analysis , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Proteome/immunology , Antibodies, Viral/blood , Antibody Specificity/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Cluster Analysis , Enzyme-Linked Immunosorbent Assay , Herpes Simplex/diagnosis , Herpes Simplex/epidemiology , Herpes Simplex/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Herpesvirus Vaccines/immunology , Humans , Protein Array Analysis/methods , Proteome/genetics , ROC Curve , Recombinant Proteins/immunology , Seroepidemiologic Studies
7.
Vaccine ; 30(3): 614-25, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22100890

ABSTRACT

Modified Vaccinia virus Ankara (MVA) is an attenuated strain of vaccinia virus that is being considered as a safer alternative to replicating vaccinia vaccine strains such as Dryvax(®) and ACAM2000. Its excellent safety profile and large genome also make it an attractive vector for the delivery of heterologous genes from other pathogens. MVA was attenuated by prolonged passage through chick embryonic fibroblasts in vitro. In human and most mammalian cells, production of infectious progeny is aborted in the late stage of infection. Despite this, MVA provides high-level gene expression and is immunogenic in humans and other animals. A key issue for vaccine developers is the ability to be able to monitor an immune response to MVA in both vaccinia naïve and previously vaccinated individuals. To this end we have used antibody profiling by proteome microarray to compare profiles before and after MVA and Dryvax vaccination to identify candidate serodiagnostic antigens. Six antigens with diagnostic utility, comprising three membrane and three non-membrane proteins from the intracellular mature virion, were purified and evaluated in ELISAs. The membrane protein WR113/D8L provided the best sensitivity and specificity of the six antigens tested for monitoring both MVA and Dryvax vaccination, whereas the A-type inclusion protein homolog, WR148, provided the best discrimination. The ratio of responses to membrane protein WR132/A13L and core protein WR070/I1L also provided good discrimination between primary and secondary responses to Dryvax, whereas membrane protein WR101/H3L and virion assembly protein WR118/D13L together provided the best sensitivity for detecting antibody in previously vaccinated individuals. These data will aid the development novel MVA-based vaccines.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral , Enzyme-Linked Immunosorbent Assay/methods , Protein Array Analysis/methods , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Humans , Recombinant Proteins , Sensitivity and Specificity , Smallpox Vaccine/administration & dosage
8.
Proteomics Clin Appl ; 5(11-12): 613-23, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956928

ABSTRACT

INTRODUCTION: Humoral immune responses play a pivotal role in naturally acquired immunity to malaria. Understanding which humoral responses are impaired among individuals at higher risk for malaria may improve our understanding of malaria immune control and contribute to vaccine development. METHODS: We compared humoral responses with 483 Plasmodium falciparum antigens between adults in, Kisumu (high, year-long malaria transmission leading to partial immunity), and adults in Kisii (low, seasonal malaria transmission). Then within each site, we compared malaria-specific humoral responses between those at higher risk for malaria (CD4(+) ≤500) and those at lower risk for malaria (CD4(+) >500). A protein microarray chip containing 483 P. falciparum antigens and 71 HIV antigens was used. Benjamini-Hochberg adjustments were made to control for multiple comparisons. RESULTS: Fifty-seven antigens including CSP, MSP1, LSA1 and AMA1 were identified as significantly more reactive in Kisumu than in Kisii. Ten of these antigens had been identified as protective in an earlier study. CD4(+) T-cell count did not significantly impact humoral responses. CONCLUSION: Protein microarrays are a useful method to screen multiple humoral responses simultaneously. This study provides useful clues for potential vaccine candidates. Modest decreases in CD4 counts may not significantly impact malaria-specific humoral immunity.


Subject(s)
HIV Infections/immunology , HIV Infections/parasitology , Immunity, Humoral , Plasmodium falciparum/immunology , Adult , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , CD4 Lymphocyte Count , Endemic Diseases/prevention & control , Female , HIV-1 , Humans , Kenya/epidemiology , Malaria Vaccines/immunology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/immunology , Male , Plasmodium falciparum/pathogenicity , Species Specificity
9.
J Proteome Res ; 10(10): 4813-24, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21863892

ABSTRACT

A complete understanding of the factors that determine selection of antigens recognized by the humoral immune response following infectious agent challenge is lacking. Here we illustrate a systems biology approach to identify the antibody signature associated with Brucella melitensis (Bm) infection in humans and predict proteomic features of serodiagnostic antigens. By taking advantage of a full proteome microarray expressing previously cloned 1406 and newly cloned 1640 Bm genes, we were able to identify 122 immunodominant antigens and 33 serodiagnostic antigens. The reactive antigens were then classified according to annotated functional features (COGs), computationally predicted features (e.g., subcellular localization, physical properties), and protein expression estimated by mass spectrometry (MS). Enrichment analyses indicated that membrane association and secretion were significant enriching features of the reactive antigens, as were proteins predicted to have a signal peptide, a single transmembrane domain, and outer membrane or periplasmic location. These features accounted for 67% of the serodiagnostic antigens. An overlay of the seroreactive antigen set with proteomic data sets generated by MS identified an additional 24%, suggesting that protein expression in bacteria is an additional determinant in the induction of Brucella-specific antibodies. This analysis indicates that one-third of the proteome contains enriching features that account for 91% of the antigens recognized, and after B. melitensis infection the immune system develops significant antibody titers against 10% of the proteins with these enriching features. This systems biology approach provides an empirical basis for understanding the breadth and specificity of the immune response to B. melitensis and a new framework for comparing the humoral responses against other microorganisms.


Subject(s)
Brucella melitensis/metabolism , Brucellosis/metabolism , Gene Expression Regulation , Antibodies/chemistry , Bacterial Proteins/chemistry , Cell Membrane/metabolism , Humans , Immune System , Lipopolysaccharides/chemistry , Mass Spectrometry/methods , Open Reading Frames , Protein Array Analysis , Proteomics/methods , Reproducibility of Results , Systems Biology
10.
Mol Cell Proteomics ; 10(10): M110.006304, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21817167

ABSTRACT

Antigen profiling using comprehensive protein microarrays is a powerful tool for characterizing the humoral immune response to infectious pathogens. Coxiella burnetii is a CDC category B bioterrorist infectious agent with worldwide distribution. In order to assess the antibody repertoire of acute and chronic Q fever patients we have constructed a protein microarray containing 93% of the proteome of Coxiella burnetii, the causative agent of Q fever. Here we report the profile of the IgG and IgM seroreactivity in 25 acute Q fever patients in longitudinal samples. We found that both early and late time points of infection have a very consistent repertoire of IgM and IgG response, with a limited number of proteins undergoing increasing or decreasing seroreactivity. We also probed a large collection of acute and chronic Q fever patient samples and identified serological markers that can differentiate between the two disease states. In this comparative analysis we confirmed the identity of numerous IgG biomarkers of acute infection, identified novel IgG biomarkers for acute and chronic infections, and profiled for the first time the IgM antibody repertoire for both acute and chronic Q fever. Using these results we were able to devise a test that can distinguish acute from chronic Q fever. These results also provide a unique perspective on isotype switch and demonstrate the utility of protein microarrays for simultaneously examining the dynamic humoral immune response against thousands of proteins from a large number of patients. The results presented here identify novel seroreactive antigens for the development of recombinant protein-based diagnostics and subunit vaccines, and provide insight into the development of the antibody response.


Subject(s)
Antigens, Bacterial/analysis , Coxiella burnetii/metabolism , Protein Array Analysis/methods , Proteome/analysis , Q Fever/immunology , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Biomarkers , Bioterrorism , Coxiella burnetii/genetics , Coxiella burnetii/immunology , Gene Expression Profiling , Humans , Immunity, Humoral/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/metabolism , Proteome/immunology
11.
Rev Sci Instrum ; 82(6): 064303, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21721711

ABSTRACT

A novel, centrifugal disk-based micro-total analysis system (µTAS) for low cost and high throughput semi-automated immunoassay processing was developed. A key innovation in the disposable immunoassay disk design is in a fluidic structure that enables very efficient micro-mixing based on a reciprocating mechanism in which centrifugal acceleration acting upon a liquid element first generates and stores pneumatic energy that is then released by a reduction of the centrifugal acceleration, resulting in a reversal of direction of flow of the liquid. Through an alternating sequence of high and low centrifugal acceleration, the system reciprocates the flow of liquid within the disk to maximize incubation/hybridization efficiency between antibodies and antigen macromolecules during the incubation/hybridization stage of the assay. The described reciprocating mechanism results in a reduction in processing time and reagent consumption by one order of magnitude.


Subject(s)
Centrifugation/instrumentation , Immunoassay/instrumentation , Microfluidic Analytical Techniques/instrumentation , Antigens, Bacterial/analysis , Antigens, Bacterial/immunology , Burkholderia , Humans , Immunoglobulin G/immunology , Indicators and Reagents/chemistry , Time Factors
12.
PLoS One ; 5(7): e11447, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20625509

ABSTRACT

BACKGROUND: Bartonella henselae is the zoonotic agent of cat scratch disease and causes potentially fatal infections in immunocompromised patients. Understanding the complex interactions between the host's immune system and bacterial pathogens is central to the field of infectious diseases and to the development of effective diagnostics and vaccines. METHODOLOGY: We report the development of a microarray comprised of proteins expressed from 96% (1433/1493) of the predicted ORFs encoded by the genome of the zoonotic pathogen Bartonella henselae. The array was probed with a collection of 62 uninfected, 62 infected, and 8 "specific-pathogen free" naïve cat sera, to profile the antibody repertoire elicited during natural Bartonella henselae infection. CONCLUSIONS: We found that 7.3% of the B. henselae proteins on the microarray were seroreactive and that seroreactivity was not evenly distributed between predicted protein function or subcellular localization. Membrane proteins were significantly most likely to be seroreactive, although only 23% of the membrane proteins were reactive. Conversely, we found that proteins involved in amino acid transport and metabolism were significantly underrepresented and did not contain any seroreactive antigens. Of all seroreactive antigens, 52 were differentially reactive with sera from infected cats, and 53 were equally reactive with sera from infected and uninfected cats. Thirteen of the seroreactive antigens were found to be differentially seroreactive between B. henselae type I and type II. Based on these results, we developed a classifier algorithm that was capable of accurately discerning 93% of the infected animals using the microarray platform. The seroreactivity and diagnostic potential of these antigens was then validated on an immunostrip platform, which correctly identified 98% of the infected cats. Our protein microarray platform provides a high-throughput, comprehensive analysis of the feline humoral immune response to natural infection with the alpha-proteobacterium B. henselae at an antigen-specific, sera-specific, and genome-wide level. Furthermore, these results provide novel insight and utility in diagnostics, vaccine development, and understanding of host-pathogen interaction.


Subject(s)
Bartonella henselae/metabolism , Bartonella henselae/pathogenicity , Immunity, Humoral/physiology , Protein Array Analysis/methods , Angiomatosis, Bacillary/immunology , Angiomatosis, Bacillary/microbiology , Animals , Bartonella Infections/immunology , Bartonella Infections/microbiology , Bartonella henselae/immunology , Cats , Immunity, Humoral/genetics , Open Reading Frames , Polymerase Chain Reaction
13.
PLoS Negl Trop Dis ; 4(5): e673, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20454614

ABSTRACT

Brucellosis is a widespread zoonotic disease that is also a potential agent of bioterrorism. Current serological assays to diagnose human brucellosis in clinical settings are based on detection of agglutinating anti-LPS antibodies. To better understand the universe of antibody responses that develop after B. melitensis infection, a protein microarray was fabricated containing 1,406 predicted B. melitensis proteins. The array was probed with sera from experimentally infected goats and naturally infected humans from an endemic region in Peru. The assay identified 18 antigens differentially recognized by infected and non-infected goats, and 13 serodiagnostic antigens that differentiate human patients proven to have acute brucellosis from syndromically similar patients. There were 31 cross-reactive antigens in healthy goats and 20 cross-reactive antigens in healthy humans. Only two of the serodiagnostic antigens and eight of the cross-reactive antigens overlap between humans and goats. Based on these results, a nitrocellulose line blot containing the human serodiagnostic antigens was fabricated and applied in a simple assay that validated the accuracy of the protein microarray results in the diagnosis of humans. These data demonstrate that an experimentally infected natural reservoir host produces a fundamentally different immune response than a naturally infected accidental human host.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Brucella melitensis/immunology , Brucellosis/immunology , Brucellosis/veterinary , Goat Diseases/immunology , Animals , Cross Reactions , Endemic Diseases/veterinary , Goats , Humans , Immunoassay/methods , Peru , Protein Array Analysis
14.
Proteomics ; 10(12): 2259-69, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20391532

ABSTRACT

Comprehensive evaluation of the humoral immune response to Coxiella burnetii may identify highly needed diagnostic antigens and potential subunit vaccine candidates. Here we report the construction of a protein microarray containing 1901 C. burnetii ORFs (84% of the entire proteome). This array was probed with Q-fever patient sera and naïve controls in order to discover C. burnetii-specific seroreactive antigens. Among the 21 seroreactive antigens identified, 13 were significantly more reactive in Q-fever cases than naïve controls. The remaining eight antigens were cross-reactive in both C. burnetii infected and naïve patient sera. An additional 64 antigens displayed variable seroreactivity in Q-fever patients, and underscore the diversity of the humoral immune response to C. burnetii. Nine of the differentially reactive antigens were validated on an alternative immunostrip platform, demonstrating proof-of-concept development of a consistent, safe, and inexpensive diagnostic assay alternative. Furthermore, we report here the identification of several new diagnostic antigens and potential subunit vaccine candidates for the highly infectious category B alphaproteobacteria, C. burnetii.


Subject(s)
Coxiella burnetii/immunology , Immunity, Humoral/genetics , Immunity, Humoral/immunology , Protein Array Analysis/methods , Q Fever/immunology , Coxiella burnetii/genetics , Gene Expression Profiling , Humans , Open Reading Frames/genetics , Polymerase Chain Reaction , Q Fever/microbiology
15.
Proc Natl Acad Sci U S A ; 106(32): 13499-504, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19666533

ABSTRACT

Understanding the way in which the immune system responds to infection is central to the development of vaccines and many diagnostics. To provide insight into this area, we fabricated a protein microarray containing 1,205 Burkholderia pseudomallei proteins, probed it with 88 melioidosis patient sera, and identified 170 reactive antigens. This subset of antigens was printed on a smaller array and probed with a collection of 747 individual sera derived from 10 patient groups including melioidosis patients from Northeast Thailand and Singapore, patients with different infections, healthy individuals from the USA, and from endemic and nonendemic regions of Thailand. We identified 49 antigens that are significantly more reactive in melioidosis patients than healthy people and patients with other types of bacterial infections. We also identified 59 cross-reactive antigens that are equally reactive among all groups, including healthy controls from the USA. Using these results we were able to devise a test that can classify melioidosis positive and negative individuals with sensitivity and specificity of 95% and 83%, respectively, a significant improvement over currently available diagnostic assays. Half of the reactive antigens contained a predicted signal peptide sequence and were classified as outer membrane, surface structures or secreted molecules, and an additional 20% were associated with pathogenicity, adaptation or chaperones. These results show that microarrays allow a more comprehensive analysis of the immune response on an antigen-specific, patient-specific, and population-specific basis, can identify serodiagnostic antigens, and contribute to a more detailed understanding of immunogenicity to this pathogen.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Burkholderia pseudomallei/immunology , Protein Array Analysis , Antigens, Bacterial/classification , Case-Control Studies , Cross Reactions/immunology , Epitope Mapping , Humans , Melioidosis/diagnosis , Melioidosis/immunology , Serologic Tests , Singapore , Thailand , United States
16.
Proteomics ; 7(10): 1678-86, 2007 May.
Article in English | MEDLINE | ID: mdl-17443847

ABSTRACT

The eradication of smallpox by vaccination with vaccinia virus was probably one of the greatest achievements of vaccinology. However, the immunological basis of this protection is not fully understood. To this end, we have used protein microarrays of the vaccinia (Western Reserve, WR) proteome to profile antibody reactivities after primary infection or boosting with the licensed smallpox vaccine, Dryvax, and with archival convalescent smallpox sera. Some 25 antigens were consistently recognized by Dryvax sera, of which half were envelope proteins (notably, H3, A13, B5, and D8). The remainder consisted mainly of core proteins (e.g. A10, L4, and I1), proteins involved in intracellular morphogenesis (A11, D13), and the A-type inclusion protein, WR148. Convalescent smallpox sera also detected vaccinia antigens on the array, consistent with the notion that there is serological cross-reactivity between these two orthopox species that underlies protection. Moreover, the profiles of immunodominant antigens recognized by variola-infected individuals and Dryvax vaccinees were indistinguishable. This is the first description of antibody-specificity profiles induced after smallpox infection. The array data indicate that a significant component of the antibody response is not involved in virus neutralization, although these antigens should be considered alongside the envelope proteins as potential candidates for diagnostic and vaccine applications.


Subject(s)
Proteome/analysis , Smallpox/blood , Vaccinia/blood , Viral Proteins/analysis , Adult , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Middle Aged , Protein Array Analysis , Smallpox/immunology , Smallpox Vaccine , Vaccination , Vaccinia/immunology , Viral Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...