Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Glycobiology ; 34(1)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-37944064

ABSTRACT

During the COVID-19 outbreak, numerous tools including protein-based vaccines have been developed. The methylotrophic yeast Pichia pastoris (synonymous to Komagataella phaffii) is an eukaryotic cost-effective and scalable system for recombinant protein production, with the advantages of an efficient secretion system and the protein folding assistance of the secretory pathway of eukaryotic cells. In a previous work, we compared the expression of SARS-CoV-2 Spike Receptor Binding Domain in P. pastoris with that in human cells. Although the size and glycosylation pattern was different between them, their protein structural and conformational features were indistinguishable. Nevertheless, since high mannose glycan extensions in proteins expressed by yeast may be the cause of a nonspecific immune recognition, we deglycosylated RBD in native conditions. This resulted in a highly pure, homogenous, properly folded and monomeric stable protein. This was confirmed by circular dichroism and tryptophan fluorescence spectra and by SEC-HPLC, which were similar to those of RBD proteins produced in yeast or human cells. Deglycosylated RBD was obtained at high yields in a single step, and it was efficient in distinguishing between SARS-CoV-2-negative and positive sera from patients. Moreover, when the deglycosylated variant was used as an immunogen, it elicited a humoral immune response ten times greater than the glycosylated form, producing antibodies with enhanced neutralizing power and eliciting a more robust cellular response. The proposed approach may be used to produce at a low cost, many antigens that require glycosylation to fold and express, but do not require glycans for recognition purposes.


Subject(s)
COVID-19 , Saccharomycetales , Vaccines , Humans , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19 Testing , Pichia/genetics , Pichia/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Recombinant Proteins/chemistry , Vaccines/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral
2.
J Appl Microbiol ; 132(6): 4277-4288, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35357068

ABSTRACT

AIMS: Bovine brucellosis is a worldwide zoonotic disease that causes important economic losses and public health concerns. Because control of the disease depends on vaccination, serodiagnosis and isolation of the infected animals, affordable, rapid and accurate point of care (POC) tests are needed. METHODS AND RESULTS: We developed and evaluated a novel glycoprotein-based immunochromatographic test for the detection of IgG antibodies against the O-polysaccharide of Brucella in bovine serum samples. Brucella GlycoStrip combines the power of immunochromatographic and bacterial glycoengineering technologies for the diagnosis of bovine brucellosis. The analysis of positive and negative reference samples indicated that the test has a diagnostic sensitivity and specificity of 96.9% (95% CI: 92.7%-100.0%) and 100%, respectively. CONCLUSIONS: Due to the recombinant glycoprotein-based antigen OAg-AcrA, which consists of the O-side chain of Brucella smooth lipopolysaccharide (sLPS) covalently linked to the carrier protein AcrA, the test is highly accurate, allows the differentiation of infected animals from those vaccinated with a rough strain or with a single dose of a smooth strain and fulfil the minimum diagnostic requirements established by the national and international regulations. SIGNIFICANCE AND IMPACT OF STUDY: This strip test could provide a rapid (10 min) and accurate diagnosis of bovine brucellosis in the field contributing to the control of the disease.


Subject(s)
Brucella , Brucellosis, Bovine , Brucellosis , Animals , Antibodies, Bacterial , Antigens, Bacterial , Brucellosis/diagnosis , Brucellosis, Bovine/diagnosis , Cattle , Enzyme-Linked Immunosorbent Assay/methods , Glycoproteins , Sensitivity and Specificity , Serologic Tests/methods , Serologic Tests/veterinary
3.
Cell Microbiol ; 22(11): e13245, 2020 11.
Article in English | MEDLINE | ID: mdl-32657513

ABSTRACT

Adhesion to host cells is the first step in the virulence cycle of any pathogen. In Gram-negative bacteria, adhesion is mediated, among other virulence factors such as the lipopolysaccharides, by specific outer-membrane proteins generally termed adhesins that belong to a wide variety of families and have different evolutionary origins. In Brucella, a widespread zoonotic pathogen of animal and human health concern, adhesion is central as it may determine the intracellular fate of the bacterium, an essential stage in its pathogenesis. In the present paper, we further characterised a genomic locus that we have previously reported encodes an adhesin (BigA) with a bacterial immunoglobulin-like domain (BIg-like). We found that this region encodes a second adhesin, which we have named BigB; and PalA, a periplasmic protein necessary for the proper display in the outer membrane of BigA and BigB. Deletion of bigB or palA diminishes the adhesion of the bacterium and overexpression of BigB dramatically increases it. Incubation of cells with the recombinant BIg-like domain of BigB induced important cytoskeletal rearrangements and affected the focal adhesion sites indicating that the adhesin targets cell-cell or cell-matrix proteins. We additionally show that PalA has a periplasmic localisation and is completely necessary for the proper display of BigA and BigB, probably avoiding their aggregation and facilitating their transport to the outer membrane. Our results indicate that this genomic island is entirely devoted to the adhesion of Brucella to host cells.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/genetics , Bacterial Proteins/metabolism , Brucella abortus/genetics , Brucella abortus/pathogenicity , Genomic Islands , Adhesins, Bacterial/genetics , Animals , Bacterial Outer Membrane/metabolism , Bacterial Proteins/genetics , Brucella abortus/physiology , Cell Line , Cell Membrane/metabolism , Cytoskeleton/metabolism , Humans , Periplasm/metabolism , Virulence
4.
J Clin Microbiol ; 58(3)2020 02 24.
Article in English | MEDLINE | ID: mdl-31826960

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) is the main cause of postdiarrheal hemolytic-uremic syndrome (HUS), a life-threatening clinical complication characterized by hemolytic anemia, thrombocytopenia, and acute renal failure that mainly affects children. A relevant feature of STEC strains is the production of Stx, and all of them express Stx1 and/or Stx2 regardless of the strain serotype. Therefore, Stx detection assays are considered the most suitable methods for the early detection of STEC infections. Single-domain antibodies from camelids (VHHs) exhibit several advantages in comparison with conventional antibodies, making them promising tools for diagnosis. In this work, we have exploited VHH technology for the development of an immunocapture assay for Stx2 detection. Thirteen anti-Stx2 VHHs previously obtained from a variable-domain repertoire library were selected and evaluated in 130 capture-detection pair combinations for Stx detection. Based on this analysis, two VHHs were selected and a double VHH-based biotin-streptavidin capture enzyme-linked immunosorbent assay (ELISA) with spectrophotometric detection was developed and optimized for Stx2 detection. This assay showed an excellent analytical and clinical sensitivity in both STEC culture supernatants and stool samples even higher than the sensitivity of a commercial ELISA. Furthermore, based on the analysis of stool samples, the VHH-based ELISA showed high correlation with stx2 detection by PCR and a commercial rapid membrane-based immunoassay. The intrinsic properties of VHHs (high target affinity and specificity, stability, and ease of expression at high yields in recombinant bacteria) and their optimal performance for Stx detection make them attractive tools for the diagnosis of HUS related to STEC (STEC-HUS).


Subject(s)
Enterohemorrhagic Escherichia coli/isolation & purification , Enzyme-Linked Immunosorbent Assay/methods , Hemolytic-Uremic Syndrome/diagnosis , Shiga Toxin 1/isolation & purification , Shiga Toxin 2/isolation & purification , Shiga-Toxigenic Escherichia coli/isolation & purification , Single-Domain Antibodies/chemistry , Animals , Argentina , Child, Preschool , Chlorocebus aethiops , Early Diagnosis , Feces/microbiology , Humans , Sensitivity and Specificity , Vero Cells
5.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31182616

ABSTRACT

The peptidoglycan in Gram-negative bacteria is a dynamic structure in constant remodeling. This dynamism, achieved through synthesis and degradation, is essential because the peptidoglycan is necessary to maintain the structure of the cell but has to have enough plasticity to allow the transport and assembly of macromolecular complexes in the periplasm and outer membrane. In addition, this remodeling has to be coordinated with the division process. Among the multiple mechanisms bacteria have to degrade the peptidoglycan are the lytic transglycosidases, enzymes of the lysozyme family that cleave the glycan chains generating gaps in the mesh structure increasing its permeability. Because these enzymes can act as autolysins, their activity has to be tightly regulated, and one of the mechanisms bacteria have evolved is the synthesis of membrane bound or periplasmic inhibitors. In the present study, we identify a periplasmic lytic transglycosidase inhibitor (PhiA) in Brucella abortus and demonstrate that it inhibits the activity of SagA, a lytic transglycosidase we have previously shown is involved in the assembly of the type IV secretion system. A phiA deletion mutant results in a strain with the incapacity to synthesize a complete lipopolysaccharide but with a higher replication rate than the wild-type parental strain, suggesting a link between peptidoglycan remodeling and speed of multiplication.


Subject(s)
Brucella abortus/pathogenicity , N-Acetylmuramoyl-L-alanine Amidase/antagonists & inhibitors , Glycoside Hydrolases/physiology , Lipopolysaccharides/biosynthesis , Multienzyme Complexes/physiology , Peptidoglycan/metabolism , Transferases/physiology , Type IV Secretion Systems/physiology , Virulence
6.
Biotechnol Bioeng ; 116(6): 1427-1438, 2019 06.
Article in English | MEDLINE | ID: mdl-30739323

ABSTRACT

In the last decades bacterial glycoengineering emerged as a new field as the result of the ability to transfer the Campylobacter jejuni N- glycosylation machinery into Escherichia coli for the production of recombinant glycoproteins that can be used as antigens for diagnosis, vaccines, and therapeutics. However, the identification of critical parameters implicated in the production process and its optimization to jump to a productive scale is still required. In this study, we developed a dual expression glycosylation vector for the production of the recombinant glycoprotein AcrA-O157, a novel antigen that allows the serodiagnosis of the infection with enterohemorrhagic E. coli O157 in humans. Volumetric productivity was studied in different culture media and found that 2xYP had 6.9-fold higher productivity than the extensively used LB. Subsequently, bioreactor batch and exponential-fed-batch cultures were designed to determine the influence of the specific growth rate (µ) on AcrA-O157 glycosylation efficiency, production kinetics, and specific productivity. At µmax , AcrA glycosylation with O157-polysaccharide and the specific synthesis rate were maximal, constituting the optimal physiological condition for AcrA-O157 production. Our findings should be considered for the design, optimization, and scaling up of AcrA-O157 production and other recombinant glycoproteins attractive for industrial applications.


Subject(s)
Bioreactors/microbiology , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Glycoproteins/metabolism , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Batch Cell Culture Techniques/methods , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/growth & development , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Glycoproteins/genetics , Glycosylation , Humans , Lipoproteins/genetics , Membrane Transport Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Pediatr Nephrol ; 34(4): 631-639, 2019 04.
Article in English | MEDLINE | ID: mdl-30367236

ABSTRACT

BACKGROUND: Providing proof of presence of Shiga toxin-producing E. coli (STEC) infection forms the basis for differentiating STEC-hemolytic uremic syndrome (HUS) and atypical HUS. As the gold standard to diagnose STEC-HUS has limitations, using ELISA to detect serum antibodies against STEC lipopolysaccharides (LPS) has proven additional value. Yet, conventional LPS-ELISA has drawbacks, most importantly presence of cross-reactivity due to the conserved lipid A part of LPS. The newly described glyco-iELISA tackles this issue by using modified LPS that eliminates the lipid A part. Here, the incremental value of glyco-iELISA compared to LPS-ELISA is assessed. METHODS: A retrospective study was performed including all pediatric patients (n = 51) presenting with a clinical pattern of STEC-HUS between 1990 and 2014 in our hospital. Subsequently, the diagnostic value of glyco-iELISA was evaluated in a retrospective nationwide study (n = 264) of patients with thrombotic microangiopathy (TMA). LPS- and glyco-iELISA were performed to detect IgM against STEC serotype O157. Both serological tests were compared with each other and with fecal diagnostics. RESULTS: Glyco-iELISA is highly sensitive and has no cross-reactivity. In the single-center cohort, fecal diagnostics, LPS-ELISA, and glyco-iELISA identified STEC O157 infection in 43%, 65%, and 78% of patients, respectively. Combining glyco-iELISA with fecal diagnostics, STEC infection due to O157 was detected in 89% of patients. In the nationwide cohort, 19 additional patients (8%) were diagnosed with STEC-HUS by glyco-iELISA. CONCLUSION: This study shows that using glyco-iELISA to detect IgM against STEC serotype O157 has clear benefit compared to conventional LPS-ELISA, contributing to optimal diagnostics in STEC-HUS.


Subject(s)
Antibodies, Bacterial/blood , Enzyme-Linked Immunosorbent Assay , Escherichia coli Infections/diagnosis , Escherichia coli O157/immunology , Hemolytic-Uremic Syndrome/diagnosis , Immunoglobulin M/blood , O Antigens/immunology , Serologic Tests , Adult , Aged , Biomarkers/blood , Escherichia coli Infections/blood , Escherichia coli Infections/microbiology , Female , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/microbiology , Humans , Male , Middle Aged , Netherlands , Pilot Projects , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Young Adult
8.
Infect Immun ; 86(11)2018 11.
Article in English | MEDLINE | ID: mdl-30126897

ABSTRACT

Brucellaceae are a group of pathogenic intracellular bacteria with the ability to modulate the host response, both at the individual cell level and systemically. One of the hallmarks of the virulence process is the capacity of the bacteria to downregulate the adaptive and acquired host immune response through a plethora of virulence factors that directly impact several key signaling cascades. PrpA is one of those virulence factors that alters, via its polyclonal B-cell activity, the humoral and cellular immune responses of the host, ultimately favoring the establishment of a chronic infection. Even though PrpA affects B cells, it directly targets macrophages, triggering a response that ultimately affects B lymphocytes. In the present article we report that PrpA is S-palmitoylated in two N-terminal cysteine residues by the host cell and that this modification is necessary for its biological activity. Our results demonstrate that S-palmitoylation promotes PrpA migration to the host cell plasma membrane and stabilizes the protein during infection. These findings add a new mechanism exploited by this highly evolved pathogen to modulate the host immune response.


Subject(s)
Brucella abortus/metabolism , Cell Membrane/metabolism , Host-Pathogen Interactions , Lipoylation , Phosphoprotein Phosphatases/metabolism , Protein Processing, Post-Translational , Animals , Cell Line , Epithelial Cells/microbiology , Humans , Immunosuppressive Agents/metabolism , Macrophages/microbiology , Mice , Protein Transport , Virulence Factors/metabolism
9.
J Infect Dis ; 217(8): 1257-1266, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29325043

ABSTRACT

Brucellaceae are stealthy pathogens with the ability to survive and replicate in the host in the context of a strong immune response. This capacity relies on several virulence factors that are able to modulate the immune system and in their structural components that have low proinflammatory activities. Lipopolysaccharide (LPS), the main component of the outer membrane, is a central virulence factor of Brucella, and it has been well established that it induces a low inflammatory response. We describe here the identification and characterization of a novel periplasmic protein (RomA) conserved in alpha-proteobacteria, which is involved in the homeostasis of the outer membrane. A mutant in this gene showed several phenotypes, such as membrane defects, altered LPS composition, reduced adhesion, and increased virulence and inflammation. We show that RomA is involved in the synthesis of LPS, probably coordinating part of the biosynthetic complex in the periplasm. Its absence alters the normal synthesis of this macromolecule and affects the homeostasis of the outer membrane, resulting in a strain with a hyperinflammatory phenotype. Our results suggest that the proper synthesis of LPS is central to maximize virulence and minimize inflammation.


Subject(s)
Bacterial Proteins/physiology , Brucella/metabolism , Brucellosis/microbiology , Lipopolysaccharides/biosynthesis , Animals , Brucella/pathogenicity , Gentamicins , Inflammation/metabolism , Mice , Protein Transport , Virulence
10.
PLoS One ; 12(10): e0182452, 2017.
Article in English | MEDLINE | ID: mdl-28981517

ABSTRACT

Shiga toxin-producing Escherichia coli (STEC) is the major etiologic agent of hemolytic-uremic syndrome (HUS). The high rate of HUS emphasizes the urgency for the implementation of primary prevention strategies to reduce its public health impact. Argentina shows the highest rate of HUS worldwide, being E. coli O157 the predominant STEC-associated HUS serogroup (>70%), followed by E. coli O145 (>9%). To specifically detect these serogroups we aimed at developing highly specific monoclonal antibodies (mAbs) against the O-polysaccharide (O-PS) section of the lipopolysaccharide (LPS) of the dominant STEC-associated HUS serogroups in Argentina. The development of hybridomas secreting mAbs against O157 or O145 was carried out through a combined immunization strategy, involving adjuvated-bacterial immunizations followed by immunizations with recombinant O-PS-protein conjugates. We selected hybridoma clones that specifically recognized the engineered O-PS-protein conjugates of O157 or O145 serogroups. Indirect ELISA of heat-killed bacteria showed specific binding to O157 or O145 serogroups, respectively, while no cross-reactivity with other epidemiological important STEC strains, Brucella abortus, Salmonella group N or Yersinia enterocolitica O9 was observed. Western blot analysis showed specific recognition of the sought O-PS section of the LPS by all mAbs. Finally, the ability of the developed mAbs to bind the surface of whole bacteria cells was confirmed by flow cytometry, confocal microscopy and agglutination assays, indicating that these mAbs present an exceptional degree of specificity and relative affinity in the detection and identification of E. coli O157 and O145 serogroups. These mAbs may be of significant value for clinical diagnosis and food quality control applications. Thus, engineered O-PS specific moieties contained in the recombinant glycoconjugates used for combined immunization and hybridoma selection are an invaluable resource for the development of highly specific mAbs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Shiga-Toxigenic Escherichia coli/immunology , Enzyme-Linked Immunosorbent Assay , Escherichia coli O157/immunology , Hybridomas , O Antigens/immunology , Serogroup , Serotyping
11.
J Am Soc Mass Spectrom ; 28(12): 2716-2723, 2017 12.
Article in English | MEDLINE | ID: mdl-28924631

ABSTRACT

Brucellaceae are Gram-negative bacteria that cause brucellosis, one of the most distributed worldwide zoonosis, transmitted to humans by contact with either infected animals or their products. The lipopolysaccharide exposed on the cell surface has been intensively studied and is considered a major virulence factor of Brucella. In the last years, structural studies allowed the determination of new structures in the core oligosaccharide and the O-antigen of this lipopolysaccharide. In this work, we have reinvestigated the lipid A structure isolated from B. suis and B. abortus lipopolysaccharides. A detailed study by MALDI-TOF mass spectrometry in the positive and negative ion modes of the lipid A moieties purified from both species was performed. Interestingly, a new feature was detected: the presence of a pyrophosphorylethanolamine residue substituting the backbone. LID-MS/MS analysis of some of the detected ions allowed assurance that the Lipid A structure composed by the diGlcN3N disaccharide, mainly hexa-acylated and penta-acylated, bearing one phosphate and one pyrophosphorylethanolamine residue. Graphical abstract ᅟ.


Subject(s)
Brucella abortus/chemistry , Brucella suis/chemistry , Lipid A/chemistry , Acylation , Brucellosis/microbiology , Disaccharides/analysis , Ethanolamines/analysis , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
12.
Vet Microbiol ; 208: 174-180, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28888634

ABSTRACT

Brucellosis is a widespread zoonotic disease caused by Brucella spp. Brucella canis is the etiological agent of canine brucellosis, a disease that can lead to sterility in bitches and dogs causing important economic losses in breeding kennels. Early and accurate diagnosis of canine brucellosis is central to control the disease and lower the risk of transmission to humans. Here, we develop and validate enzyme and lateral flow immunoassays for improved serodiagnosis of canine brucellosis using as antigen the B. canis rough lipopolysaccharide (rLPS). The method used to obtain the rLPS allowed us to produce more homogeneous batches of the antigen that facilitated the standardization of the assays. To validate the assays, 284 serum samples obtained from naturally infected dogs and healthy animals were analyzed. For the B. canis-iELISA and B. canis-LFIA the diagnostic sensitivity was of 98.6%, and the specificity 99.5% and 100%, respectively. We propose the implementation of the B. canis-LFIA as a screening test in combination with the highly accurate laboratory g-iELISA. The B. canis-LFIA is a rapid, accurate and easy to use test, characteristics that make it ideal for the serological surveillance of canine brucellosis in the field or veterinary laboratories. Finally, a blind study including 1040 serum samples obtained from urban dogs showed a prevalence higher than 5% highlighting the need of new diagnostic tools for a more effective control of the disease in dogs and therefore to reduce the risk of transmission of this zoonotic pathogen to humans.


Subject(s)
Brucellosis/veterinary , Dog Diseases/diagnosis , Immunoassay/veterinary , Animals , Argentina/epidemiology , Brucellosis/diagnosis , Brucellosis/epidemiology , Brucellosis/microbiology , Dog Diseases/epidemiology , Dog Diseases/microbiology , Dogs , Immunoassay/methods , Reproducibility of Results , Sensitivity and Specificity
13.
J Clin Microbiol ; 54(6): 1448-1455, 2016 06.
Article in English | MEDLINE | ID: mdl-26984975

ABSTRACT

Brucellosis is a highly zoonotic disease that affects animals and human beings. Brucella suis is the etiological agent of porcine brucellosis and one of the major human brucellosis pathogens. Laboratory diagnosis of porcine brucellosis mainly relies on serological tests, and it has been widely demonstrated that serological assays based on the detection of anti O-polysaccharide antibodies are the most sensitive tests. Here, we validate a recombinant glycoprotein antigen, an N-formylperosamine O-polysaccharide-protein conjugate (OAg-AcrA), for diagnosis of porcine brucellosis. An indirect immunoassay based on the detection of anti-O-polysaccharide IgG antibodies was developed coupling OAg-AcrA to enzyme-linked immunosorbent assay plates (glyco-iELISA). To validate the assay, 563 serum samples obtained from experimentally infected and immunized pigs, as well as animals naturally infected with B. suis biovar 1 or 2, were tested. A receiver operating characteristic (ROC) analysis was performed, and based on this analysis, the optimum cutoff value was 0.56 (relative reactivity), which resulted in a diagnostic sensitivity and specificity of 100% and 99.7%, respectively. A cutoff value of 0.78 resulted in a test sensitivity of 98.4% and a test specificity of 100%. Overall, our results demonstrate that the glyco-iELISA is highly accurate for diagnosis of porcine brucellosis, improving the diagnostic performance of current serological tests. The recombinant glycoprotein OAg-AcrA can be produced in large homogeneous batches in a standardized way, making it an ideal candidate for further validation as a universal antigen for diagnosis of "smooth" brucellosis in animals and humans.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Brucella/immunology , Brucellosis/diagnosis , Serologic Tests/methods , Swine Diseases/diagnosis , Animals , Antigens, Bacterial/genetics , Female , Male , ROC Curve , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sensitivity and Specificity , Swine
14.
Biosens Bioelectron ; 80: 24-33, 2016 Jun 15.
Article in English | MEDLINE | ID: mdl-26802749

ABSTRACT

Access to appropriate diagnostic tools is an essential component in the evaluation and improvement of global health. Additionally, timely detection of infectious agents is critical in early diagnosis and treatment of infectious diseases. Conventional pathogen detection methods such as culturing, enzyme linked immunosorbent assay (ELISA) or polymerase chain reaction (PCR) require long assay times, and complex and expensive instruments making them not adaptable to point-of-care (PoC) needs at resource-constrained places and primary care settings. Therefore, there is an unmet need to develop portable, simple, rapid, and accurate methods for PoC detection of infections. Here, we present the development and validation of a portable, robust and inexpensive electrochemical magnetic microbeads-based biosensor (EMBIA) platform for PoC serodiagnosis of infectious diseases caused by different types of microorganisms (parasitic protozoa, bacteria and viruses). We demonstrate the potential use of the EMBIA platform for in situ diagnosis of human (Chagas disease and human brucellosis) and animal (bovine brucellosis and foot-and-mouth disease) infections clearly differentiating infected from non-infected individuals or animals. For Chagas disease, a more extensive validation of the test was performed showing that the EMBIA platform displayed an excellent diagnostic performance almost indistinguishable, in terms of specificity and sensitivity, from a fluorescent immunomagnetic assay and the conventional ELISA using the same combination of antigens. This platform technology could potentially be applicable to diagnose other infectious and non-infectious diseases as well as detection and/or quantification of biomarkers at the POC and primary care settings.


Subject(s)
Biosensing Techniques , Communicable Diseases/blood , Enzyme-Linked Immunosorbent Assay/methods , Serologic Tests/methods , Animals , Bacteria/isolation & purification , Bacteria/pathogenicity , Communicable Diseases/microbiology , Communicable Diseases/parasitology , Communicable Diseases/virology , Humans , Magnetics , Parasites/isolation & purification , Parasites/pathogenicity , Point-of-Care Systems , Viruses/isolation & purification , Viruses/pathogenicity
15.
J Clin Microbiol ; 53(2): 528-38, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25472487

ABSTRACT

Human infection with Shiga toxin-producing Escherichia coli (STEC) is a major cause of postdiarrheal hemolytic-uremic syndrome (HUS), a life-threatening condition characterized by hemolytic anemia, thrombocytopenia, and acute renal failure. E. coli O157:H7 is the dominant STEC serotype associated with HUS worldwide, although non-O157 STEC serogroups can cause a similar disease. The detection of anti-O157 E. coli lipopolysaccharide (LPS) antibodies in combination with stool culture and detection of free fecal Shiga toxin considerably improves the diagnosis of STEC infections. In the present study, we exploited a bacterial glycoengineering technology to develop recombinant glycoproteins consisting of the O157, O145, or O121 polysaccharide attached to a carrier protein as serogroup-specific antigens for the serological diagnosis of STEC-associated HUS. Our results demonstrate that using these antigens in indirect ELISAs (glyco-iELISAs), it is possible to clearly discriminate between STEC O157-, O145-, and O121-infected patients and healthy children, as well as to confirm the diagnosis in HUS patients for whom the classical diagnostic procedures failed. Interestingly, a specific IgM response was detected in almost all the analyzed samples, indicating that it is possible to detect the infection in the early stages of the disease. Additionally, in all the culture-positive HUS patients, the serotype identified by glyco-iELISAs was in accordance with the serotype of the isolated strain, indicating that these antigens are valuable not only for diagnosing HUS caused by the O157, O145, and O121 serogroups but also for serotyping and guiding the subsequent steps to confirm diagnosis.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Glycoproteins/immunology , Hemolytic-Uremic Syndrome/diagnosis , Serotyping/methods , Shiga-Toxigenic Escherichia coli/immunology , Antigens, Bacterial/genetics , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay/methods , Glycoproteins/genetics , Humans , Immunoglobulin M/blood , Infant , Infant, Newborn , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Retrospective Studies , Single-Blind Method
16.
Vet Microbiol ; 172(3-4): 455-65, 2014 Aug 27.
Article in English | MEDLINE | ID: mdl-24984948

ABSTRACT

Brucellosis is a highly contagious zoonosis that affects livestock and human beings. Laboratory diagnosis of bovine brucellosis mainly relies on serological diagnosis using serum and/or milk samples. Although there are several serological tests with different diagnostic performance and capacity to differentiate vaccinated from infected animals, there is still no standardized reference antigen for the disease. Here we validate the first recombinant glycoprotein antigen, an N-formylperosamine O-polysaccharide-protein conjugate (OAg-AcrA), for diagnosis of bovine brucellosis. This antigen can be produced in homogeneous batches without the need of culturing pathogenic brucellae; all characteristics that make it appropriate for standardization. An indirect immunoassay based on the detection of anti O-polysaccharide IgG antibodies in bovine samples was developed coupling OAg-AcrA to magnetic beads or ELISA plates. As a proof of concept and to validate the antigen, we analyzed serum, whole blood and milk samples obtained from non-infected, experimentally infected and vaccinated animals included in a vaccination/infection trial performed in our laboratory as well as more than 1000 serum and milk samples obtained from naturally infected and S19-vaccinated animals from Argentina. Our results demonstrate that OAg-AcrA-based assays are highly accurate for diagnosis of bovine brucellosis, even in vaccinated herds, using different types of samples and in different platforms. We propose this novel recombinant glycoprotein as an antigen suitable for the development of new standard immunological tests for screening and confirmatory diagnosis of bovine brucellosis in regions or countries with brucellosis-control programs.


Subject(s)
Antigens, Bacterial/immunology , Brucella/immunology , Brucellosis, Bovine/diagnosis , Glycoproteins/immunology , Animals , Bacterial Vaccines/immunology , Brucellosis, Bovine/prevention & control , Cattle , Fluorescent Antibody Technique, Indirect/methods , Fluorescent Antibody Technique, Indirect/veterinary , Humans , Milk/immunology , Milk/virology , Protein Engineering , Recombinant Proteins , Reproducibility of Results , Serologic Tests/veterinary
17.
Microb Pathog ; 67-68: 8-13, 2014.
Article in English | MEDLINE | ID: mdl-24508400

ABSTRACT

Brucellosis, a disease caused by the gram-negative bacterium Brucella spp., is a widespread zoonosis that inflicts important animal and human health problems, especially in developing countries. One of the hallmarks of Brucella infection is its capacity to establish a chronic infection, characteristic that depends on a wide repertoire of virulence factors among which are immunomodulatory proteins such as PrpA (encoding the proline racemase protein A or hydroxyproline-2-epimerase), involved in the establishment of the chronic phase of the infectious process that we have previously identified and characterized. We report here that, in vivo, Brucella abortus prpA is responsible for an increment in the B-cell number and in the specific antibody response and that these antibodies promote cell infection. We additionally found that Brucella alters the cytokine levels of IFN-γ, IL-10, TGFß1 and TNFα during the acute phase of the infectious process in a prpA dependent manner.


Subject(s)
Amino Acid Isomerases/immunology , Bacterial Proteins/immunology , Brucella abortus/enzymology , Brucellosis/immunology , Brucellosis/microbiology , Amino Acid Isomerases/genetics , Animals , Antibodies, Bacterial/immunology , B-Lymphocytes/immunology , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucella abortus/immunology , Brucellosis/genetics , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virulence Factors/genetics , Virulence Factors/immunology
18.
Cell Microbiol ; 16(3): 396-410, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24119283

ABSTRACT

Brucella abortus, the aetiological agent of bovine brucellosis, is an intracellular pathogen whose virulence is completely dependent on a type IV secretion system. This secretion system translocates effector proteins into the host cell to modulate the intracellular fate of the bacterium in order to establish a secure niche were it actively replicates. Although much has been done in understanding how this secretion system participates in the virulence process, few effector proteins have been identified to date. We describe here the identification of a type IV secretion substrate (SepA) that is only present in Brucella spp. and has no detectable homology to known proteins. This protein is secreted in a virB-dependent manner in a two-step process involving a periplasmic intermediate and secretion is necessary for its function. The deletion mutant showed a defect in the early stages of intracellular replication in professional and non-professional phagocytes although it invades the cells more efficiently than the wild-type parental strain. Our results indicate that, even though the mutant was more invasive, it had a defect in excluding the lysosomal marker Lamp-1 and was inactivated more efficiently during the early phases of the intracellular life cycle.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Brucella abortus/pathogenicity , Animals , Bacterial Secretion Systems , Brucella abortus/genetics , Brucellosis/microbiology , Brucellosis/pathology , Cell Line , Female , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Lysosomal Membrane Proteins , Mice , Mice, Inbred BALB C , Virulence Factors/metabolism
19.
J Biol Chem ; 288(28): 20208-16, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720774

ABSTRACT

Brucella spp. and Trypanosoma cruzi are two intracellular pathogens that have no evolutionary common origins but share a similar lifestyle as they establish chronic infections for which they have to circumvent the host immune response. Both pathogens have a virulence factor (prpA in Brucella and tcPrac in T. cruzi) that induces B-cell proliferation and promotes the establishment of the chronic phase of the infectious process. We show here that, even though PrpA promotes B-cell proliferation, it targets macrophages in vitro and is translocated to the cytoplasm during the intracellular replication phase. We observed that PrpA-treated macrophages induce the secretion of a soluble factor responsible for B-cell proliferation and identified nonmuscular myosin IIA (NMM-IIA) as a receptor required for binding and function of this virulence factor. Finally, we show that the Trypanosoma cruzi homologue of PrpA also targets macrophages to induce B-cell proliferation through the same receptor, indicating that this virulence strategy is conserved between a bacterial and a protozoan pathogen.


Subject(s)
B-Lymphocytes/immunology , Bacterial Proteins/immunology , Cell Proliferation , Macrophages/immunology , Virulence Factors/immunology , Amino Acid Isomerases/genetics , Amino Acid Isomerases/immunology , Amino Acid Isomerases/metabolism , Animals , B-Lymphocytes/cytology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Brucella abortus/immunology , Brucella abortus/metabolism , Brucella abortus/pathogenicity , Cell Line , Cells, Cultured , Female , Macrophages/parasitology , Macrophages/virology , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Nonmuscle Myosin Type IIA/immunology , Nonmuscle Myosin Type IIA/metabolism , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Trypanosoma cruzi/immunology , Trypanosoma cruzi/metabolism , Trypanosoma cruzi/pathogenicity , Virulence/immunology , Virulence Factors/genetics , Virulence Factors/metabolism
20.
PLoS Negl Trop Dis ; 7(2): e2048, 2013.
Article in English | MEDLINE | ID: mdl-23459192

ABSTRACT

Brucellosis is a highly contagious zoonosis and still a major human health problem in endemic areas of the world. Although several diagnostic tools are available, most of them are difficult to implement especially in developing countries where complex health facilities are limited. Taking advantage of the identical structure and composition of the Brucella spp. and Yersinia enterocolitica O:9 O-polysaccharide, we explored the application of a recombinant Y. enterocolitica O:9-polysaccharide-protein conjugate (OAg-AcrA) as a novel antigen for diagnosis of human brucellosis. We have developed and validated an indirect immunoassay using OAg-AcrA coupled to magnetic beads. OAg-AcrA was produced and purified with high yields in Y. enterocolitica O:9 cells co-expressing the oligosaccharyltransferase PglB and the protein acceptor AcrA of Campylobacter jejuni without the need for culturing Brucella. Expression of PglB and AcrA in Y. enterocolitica resulted in the transfer of the host O-polysaccharide from its lipid carrier to AcrA. To validate the assay and determine the cutoff values, a receiver-operating characteristic analysis was performed using a panel of characterized serum samples obtained from healthy individuals and patients of different clinical groups. Our results indicate that, using this assay, it is possible to detect infection caused by the three main human brucellosis agents (B. abortus, B. melitensis and B. suis) and select different cutoff points to adjust sensitivity and specificity levels as needed. A cutoff value of 13.20% gave a sensitivity of 100% and a specificity of 98.57%, and a cutoff value of 16.15% resulted in a test sensitivity and specificity of 93.48% and 100%, respectively. The high diagnostic accuracy, low cost, reduced assay time and simplicity of this new glycoconjugate-magnetic beads assay makes it an attractive diagnostic tool for using not only in clinics and brucellosis reference laboratories but also in locations with limited laboratory infrastructure and/or minimally trained community health workers.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial , Brucellosis/diagnosis , Diagnostic Tests, Routine/methods , Magnetics , Microspheres , Humans , Immunoassay/methods , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...