Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 837
Filtrar
1.
Biotechnol Prog ; 39(3): e3333, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36795072

RESUMEN

In good manufacturing practice (GMP) facilities in the biopharmaceutical industry, chromatography resins are largely underutilized during purification of single drug products during clinical production. Chromatography resins are dedicated to a specific product and disposed of, after only a fraction of their lifetime due to concerns of potential product carryover from one program to another. In this study, we follow a resin lifetime methodology typically used for commercial submissions and apply it to determine the feasibility of purifying different products on a Protein A MabSelect PrismA™ resin. Three distinct monoclonal antibodies were used as model molecules. Column performance was monitored through chromatogram profiles, yield, clearance capability of selected media components, pressure and product quality. A protein carryover study was designed to demonstrate that the column cleaning procedures reduced protein carryover to safe cleanliness levels regardless of multiple product contact cycles and the order in which the mAbs are captured. Data show that up to 90 total cycles (30 cycles per antibody), there was negligible protein carryover and impact on process performance. Product quality was consistent, with the only meaningful trends found for the leached Protein A ligand, without affecting the conclusion of the study. While the study was restricted to three antibodies, the proof of concept for resin reuse was demonstrated.


Asunto(s)
Anticuerpos Monoclonales , Cromatografía de Afinidad , Equipo Reutilizado , Proteína Estafilocócica A , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Cromatografía de Afinidad/instrumentación , Cromatografía de Afinidad/métodos , Estudios de Factibilidad , Inmunoglobulina G/inmunología , Proteína Estafilocócica A/inmunología , Ligandos , Medición de Riesgo
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35058363

RESUMEN

Gram-positive organisms with their thick envelope cannot be lysed by complement alone. Nonetheless, antibody-binding on the surface can recruit complement and mark these invaders for uptake and killing by phagocytes, a process known as opsonophagocytosis. The crystallizable fragment of immunoglobulins (Fcγ) is key for complement recruitment. The cell surface of S. aureus is coated with Staphylococcal protein A (SpA). SpA captures the Fcγ domain of IgG and interferes with opsonization by anti-S. aureus antibodies. In principle, the Fcγ domain of therapeutic antibodies could be engineered to avoid the inhibitory activity of SpA. However, the SpA-binding site on Fcγ overlaps with that of the neonatal Fc receptor (FcRn), an interaction that is critical for prolonging the half-life of serum IgG. This evolutionary adaptation poses a challenge for the exploration of Fcγ mutants that can both weaken SpA-IgG interactions and retain stability. Here, we use both wild-type and transgenic human FcRn mice to identify antibodies with enhanced half-life and increased opsonophagocytic killing in models of S. aureus infection and demonstrate that antibody-based immunotherapy can be improved by modifying Fcγ. Our experiments also show that by competing for FcRn-binding, staphylococci effectively reduce the half-life of antibodies during infection. These observations may have profound impact in treating cancer, autoimmune, and asthma patients colonized or infected with S. aureus and undergoing monoclonal antibody treatment.


Asunto(s)
Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Opsonización/inmunología , Ingeniería de Proteínas , Secuencia de Aminoácidos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Activación de Complemento , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta Inmunológica , Humanos , Fagocitosis/inmunología , Unión Proteica , Ingeniería de Proteínas/métodos , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/inmunología , Receptores Fc/genética , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología
3.
Anal Biochem ; 641: 114403, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34610335

RESUMEN

The Cry1Ab toxin is usually expressed in genetically modified crops in order to control chewing pests. Although the gold immunochromatography assay (GICA) based on the double-antibody sandwich method has been developed to detect this toxin, its detection sensitivity needs improvement. In this study, Cry1Ab-51 antibodies were immobilized orientationally in a simple and effective way on colloidal gold nanoparticles (CGNPs) using the affinity of staphylococcal protein A (SPA) towards the fragment crystallizable (FC) fragment of mouse immunoglobulin G (IgG). Lateral flow detection test strips, assembled with probes labeled with orientational methods under optimal operational conditions (new probe), were 10 times more sensitive than test strips assembled with probes labeled by adsorption (conventional probe). Experiments showed that the affinity of the new probe was much higher than the conventional probe. The immunochromatography gold strip (ICG strip) assembled using the new probe was highly specific to Cry1Ab with no cross-reaction with other transgenic proteins, and it was proved that the specificity of the new probe had no change. Furthermore, the ICG strips assembled with the new probe could be stored for 12 months under dry conditions without a significant loss of sensitivity. The orientational labeling of the antibodies with SPA on colloidal gold proved to be suitable for improving the sensitivity of the ICG strips.


Asunto(s)
Anticuerpos Monoclonales/análisis , Oro/química , Nanopartículas del Metal/química , Proteína Estafilocócica A/química , Anticuerpos Monoclonales/inmunología , Cromatografía de Afinidad , Proteína Estafilocócica A/inmunología
4.
Microb Cell Fact ; 20(1): 212, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789248

RESUMEN

Protein A (SpA) is one of the most important Staphylococcus aureus cell wall proteins. It includes five immunoglobulin (Ig)-binding domains which can bind to immune complexes through the Fc region of immunoglobulins. The binding of SpA to the polymeric supports can be used to prepare affinity chromatography resins, which are useful for immunoprecipitation (IP) of antibodies. Protein A is also used to purify many anti-cancer antibodies. In this study, SpA was displayed on the surface of Bacillus subtilis cells using a sortase-mediated system to display the target protein to the B. subtilis cell wall. A series of plasmids consisting of cassettes for cell wall-directed protein A as well as negative controls were constructed and transformed into B. subtilis WASD (wprA sigD) cells. SDS-PAGE, western blot, flow cytometry, functional IgG purification assay, and a modified ELISA assay were used to confirm the surface display of SpA and evaluate its function. Semi-quantitative ELISA results showed that the binding capacity of lyophilized Bs-SpA is 100 µg IgG from rabbit serum per 1 mg of cells under optimal experimental conditions. Low production costs, optimal performance, and the use of a harmless strain compared to a similar commercial product predict the possible use of SpA immobilization technology in the future.


Asunto(s)
Aminoaciltransferasas/metabolismo , Bacillus subtilis/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteína Estafilocócica A/metabolismo , Anticuerpos/química , Anticuerpos/aislamiento & purificación , Bacillus subtilis/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Pared Celular/enzimología , Pared Celular/metabolismo , Unión Proteica , Proteína Estafilocócica A/química , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/química
5.
Parasit Vectors ; 14(1): 571, 2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34749796

RESUMEN

BACKGROUND: Vaccines against the sexual stages of the malarial parasite Plasmodium falciparum are indispensable for controlling malaria and abrogating the spread of drug-resistant parasites. Pfs25, a surface antigen of the sexual stage of P. falciparum, is a leading candidate for transmission-blocking vaccine development. While clinical trials have reported that Pfs25-based vaccines are safe and effective in inducing transmission-blocking antibodies, the extent of the genetic diversity of Pfs25 in malaria endemic populations has rarely been studied. Thus, this study aimed to investigate the global diversity of Pfs25 in P. falciparum populations. METHODS: A database of 307 Pfs25 sequences of P. falciparum was established. Population genetic analyses were performed to evaluate haplotype and nucleotide diversity, analyze haplotypic distribution patterns of Pfs25 in different geographical populations, and construct a haplotype network. Neutrality tests were conducted to determine evidence of natural selection. Homology models of the Pfs25 haplotypes were constructed, subjected to molecular dynamics (MD), and analyzed in terms of flexibility and percentages of secondary structures. RESULTS: The Pfs25 gene of P. falciparum was found to have 11 unique haplotypes. Of these, haplotype 1 (H1) and H2, the major haplotypes, represented 70% and 22% of the population, respectively, and were dominant in Asia, whereas only H1 was dominant in Africa, Central America, and South America. Other haplotypes were rare and region-specific, resulting in unique distribution patterns in different geographical populations. The diversity in Pfs25 originated from ten single-nucleotide polymorphism (SNP) loci located in the epidermal growth factor (EGF)-like domains and anchor domain. Of these, an SNP at position 392 (GGA/GCA), resulting in amino acid substitution 131 (Gly/Ala), defined the two major haplotypes. The MD results showed that the structures of H1 and H2 variants were relatively similar. Limited polymorphism in Pfs25 could likely be due to negative selection. CONCLUSIONS: The study successfully established a Pfs25 sequence database that can become an essential tool for monitoring vaccine efficacy, designing assays for detecting malaria carriers, and conducting epidemiological studies of P. falciparum. The discovery of the two major haplotypes, H1 and H2, and their conserved structures suggests that the current Pfs25-based vaccines could be used globally for malaria control.


Asunto(s)
Antígenos de Protozoos/genética , Vacunas contra la Malaria/genética , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteína Estafilocócica A/genética , Antígenos de Protozoos/inmunología , Variación Genética , Haplotipos , Humanos , Vacunas contra la Malaria/inmunología , Malaria Falciparum/transmisión , Plasmodium falciparum/inmunología , Polimorfismo de Nucleótido Simple , Proteínas Protozoarias/inmunología , Proteína Estafilocócica A/inmunología
6.
Front Immunol ; 12: 650782, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367127

RESUMEN

The changes in the serum levels of aquaporin-4-IgG (AQP4-IgG), immunoglobulins, and inflammatory mediators in neuromyelitis optica spectrum disorder (NMOSD) cases treated with immunoadsorption have been rarely described in detail. Here we report a 29-year-old steroid-resistant NMOSD female with a severe disability (bilateral blindness and paraplegia) who received protein-A immunoadsorption as a rescue treatment. During the total 5 sessions, the circulating level of AQP4-IgG, immunoglobulins, and complement proteins (C3 and C4) showed a rapid and sawtooth-like decrease, and the serum AQP4-IgG titer declined from 1:320 to below the detectable limit at the end of the 3rd procedure. Of all the antibodies, IgG had the biggest removal rate (>96.1%), followed by IgM (>66.7%) and IgA (53%), while complement C3 and C4 also dropped by 73% and 65%, respectively. The reduced pro-inflammatory cytokines (interleukin-8 and tumor necrosis factor-α) and marked increased lymphocyte (T and B cell) counts were also observed. The improvement of symptoms initiated after the last session, with a low AQP4-IgG titer (1:32) persisting thereafter. Accordingly, protein-A immunoadsorption treatment could be one of the potential rescue therapies for steroid-resistant NMOSD patients with a severe disability.


Asunto(s)
Acuaporina 4/inmunología , Biomarcadores/sangre , Inmunoglobulina G/inmunología , Neuromielitis Óptica/terapia , Plasmaféresis/métodos , Proteína Estafilocócica A/inmunología , Adulto , Complemento C3/inmunología , Complemento C4/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Interleucina-8 , Recuento de Linfocitos , Neuromielitis Óptica/sangre , Neuromielitis Óptica/inmunología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología
7.
Protein Expr Purif ; 186: 105930, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34147604

RESUMEN

Diamond MMC Mustang is a relatively new mixed-mode resin, which mediates both cation exchange and hydrophobic interactions. In this work, we evaluated this resin using Cytiva's Capto MMC ImpRes, a well-established mixed-mode resin with similar properties, as a benchmark. The data suggest that in comparison with Capto MMC ImpRes, Diamond MMC Mustang exhibits comparable binding capacity and resolution. In addition, the resin under evaluation shows good lot-to-lot consistency. The information provided in this study allows users to have additional options when selecting mixed-mode resin for intermediate purification or final polishing, which is favourable especially at the present time when the supply chains of many manufacturers are negatively impacted by the coronavirus pandemic.


Asunto(s)
Anticuerpos Biespecíficos/aislamiento & purificación , Cromatografía de Afinidad/métodos , Animales , Anticuerpos Biespecíficos/inmunología , Benchmarking , Células CHO , Cromatografía de Afinidad/instrumentación , Cricetulus , Proteína Estafilocócica A/análisis , Proteína Estafilocócica A/inmunología
8.
mBio ; 12(3): e0089921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34060329

RESUMEN

One of the defining features of Staphylococcus aureus is its ability to evade and impair the human immune response through expression of staphylococcal protein A (SpA). Herein, we describe a previously unknown mechanism by which SpA can form toxic immune complexes when in the presence of human serum, which leads to the loss of human leukocytes. Further, we demonstrate that these toxic complexes are formed specifically through SpA's interaction with intact human IgG and that, in the presence of purified IgG Fab and Fc fragments, SpA shows no such toxicity. The mechanism of action of this toxicity appears to be one mediated by necrosis and not by apoptosis, as previously hypothesized, with up to 90% of human B cells rapidly becoming necrotic following stimulation with SpA-IgG complexes. This phenomenon depends on the immunoglobulin binding capacity of SpA, as a nonbinding mutant of SpA did not induce necrosis. Importantly, immune sera raised against SpA had the capacity to significantly reduce the observed toxicity. An unprecedented toxic effect of SpA-IgG complexes on monocytes was also observed, suggesting the existence of a novel mechanism independent from the interaction of SpA with the B cell receptor. Together, these data implicate SpA in inducing indiscriminate leukocyte toxicity upon formation of complexes with IgG and highlight the requirement for vaccination strategies to inhibit this mechanism. IMPORTANCE Staphylococcus aureus is one of the largest health care threats faced by humankind, with a reported mortality rate within the United States greater than that of HIV/AIDS, tuberculosis, and viral hepatitis combined. One of the defining features of S. aureus as a human pathogen is its ability to evade and impair the human immune response through expression of staphylococcal protein A. Herein, we show that SpA induces necrosis in various immune cells by complexing with human immunoglobulins. Vaccination of mice with a nontoxigenic SpA mutant induced sera capable of inhibiting this mechanism. These observations shed new light on the toxic mechanisms of this key staphylococcal virulence factor and on protective modalities of SpA-based vaccination.


Asunto(s)
Complejo Antígeno-Anticuerpo , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Inmunoglobulina G/metabolismo , Necrosis/inmunología , Proteína Estafilocócica A/farmacología , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos BALB C , Proteína Estafilocócica A/administración & dosificación , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/metabolismo , Vacunación
9.
Front Immunol ; 12: 662782, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995388

RESUMEN

Staphylococcus aureus, a common cause of serious and often fatal infections, is well-armed with secreted factors that disarm host immune defenses. Highly expressed in vivo during infection, Staphylococcal protein A (SpA) is reported to also contribute to nasal colonization that can be a prelude to invasive infection. Co-evolution with the host immune system has provided SpA with an Fc-antibody binding site, and a Fab-binding site responsible for non-immune superantigen interactions via germline-encoded surfaces expressed on many human BCRs. We wondered whether the recurrent exposures to S. aureus commonly experienced by adults, result in the accumulation of memory B-cell responses to other determinants on SpA. We therefore isolated SpA-specific class-switched memory B cells, and characterized their encoding VH : VL antibody genes. In SpA-reactive memory B cells, we confirmed a striking bias in usage for VH genes, which retain the surface that mediates the SpA-superantigen interaction. We postulate these interactions reflect co-evolution of the host immune system and SpA, which during infection results in immune recruitment of an extraordinarily high prevalence of B cells in the repertoire that subverts the augmentation of protective defenses. Herein, we provide the first evidence that human memory responses are supplemented by B-cell clones, and circulating-antibodies, that bind to SpA determinants independent of the non-immune Fc- and Fab-binding sites. In parallel, we demonstrate that healthy individuals, and patients recovering from S. aureus infection, both have circulating antibodies with these conventional binding specificities. These findings rationalize the potential utility of incorporating specially engineered SpA proteins into a protective vaccine.


Asunto(s)
Linfocitos B/inmunología , Evolución Clonal/inmunología , Memoria Inmunológica , Proteína Estafilocócica A/inmunología , Linfocitos B/metabolismo , Biomarcadores , Humanos , Inmunofenotipificación , Modelos Biológicos , Unión Proteica/inmunología , Conformación Proteica , Proteína Estafilocócica A/química , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/inmunología , Relación Estructura-Actividad , Superantígenos/inmunología
10.
mBio ; 12(2)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879590

RESUMEN

Staphylococcus aureus causes reiterative and chronic persistent infections. This can be explained by the formidable ability of this pathogen to escape immune surveillance mechanisms. Cells of S. aureus display the abundant staphylococcal protein A (SpA). SpA binds to immunoglobulin (Ig) molecules and coats the bacterial surface to prevent phagocytic uptake. SpA also binds and cross-links variable heavy 3 (VH3) idiotype (IgM) B cell receptors, promoting B cell expansion and the secretion of nonspecific VH3-IgM via a mechanism requiring CD4+ T cell help. SpA binding to antibodies is mediated by the N-terminal Ig-binding domains (IgBDs). The so-called region X, uncharacterized LysM domain, and C-terminal LPXTG sorting signal for peptidoglycan attachment complete the linear structure of the protein. Here, we report that both the LysM domain and the LPXTG motif sorting signal are required for the B cell superantigen activity of SpA in a mouse model of infection. SpA molecules purified from staphylococcal cultures are sufficient to exert B cell superantigen activity and promote immunoglobulin secretion as long as they carry intact LysM and LPXTG motif domains with bound peptidoglycan fragments. The LysM domain binds the glycan chains of peptidoglycan fragments, whereas the LPXTG motif is covalently linked to wall peptides lacking glycan. These findings emphasize the complexity of SpA interactions with B cell receptors.IMPORTANCE The LysM domain is found in all kingdoms of life. While their function in mammals is not known, LysM domains of bacteria and their phage parasites are associated with enzymes that cleave or remodel peptidoglycan. Plants recognize microbe-associated molecular patterns such as chitin via receptors endowed with LysM-containing ectodomains. In plants, such receptors play equally important roles in defense and symbiosis signaling. SpA of S. aureus carries a LysM domain that binds glycan strands of peptidoglycan to influence defined B cell responses that divert pathogen-specific adaptive immune responses.


Asunto(s)
Linfocitos B/inmunología , Peptidoglicano/inmunología , Peptidoglicano/metabolismo , Proteína Estafilocócica A/inmunología , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Inmunidad Adaptativa , Animales , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Polisacáridos/inmunología , Polisacáridos/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos B , Proteína Estafilocócica A/genética
11.
Protein Expr Purif ; 177: 105764, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32971296

RESUMEN

Immobilizing antibodies on the nitrocellulose membrane is an important step to increase the sensitivity of the Lateral Flow Test strip for detecting pathogenic antigen. In our research, the fusion protein between nitrocellulose-binding anchor protein 3-Helix - a protein that has a strong affinity to nitrocellulose membrane and protein A - a protein that can bind to the Fc tail of IgG antibody was generated. This fusion protein was expected to help IgG antibodies to be more strongly binding and oriented immobilized onto the nitrocellulose membrane. The recombinant vector pET22b-proA and pET22b-proA-3-Helix coded for protein A and protein A-3-Helix were cloned. These proteins were overexpressed in BL21 and purified by immobilized metal affinity chromatography with purity above 90%. The purified protein was used to evaluate the orientation binding on nitrocellulose membranes by lateral flow challenge. Results showed that protein A-3-Helix binding to nitrocellulose membrane was better than that of protein A. The former protein increased antibody binding and stereochemical immobilizing onto nitrocellulose membrane compared to its protein A counterpart. In summary, we have succeeded in cloning, purifying, and characterizing a dual-head recombinant protein A and protein A-3-Helix. The results show the potential application of protein A-3-Helix in the immobilizing antibody on the test strip.


Asunto(s)
Cromatografía de Afinidad/métodos , Colodión/química , Proteínas Inmovilizadas/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Proteína Estafilocócica A/química , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/inmunología , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Unión Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteína Estafilocócica A/genética , Proteína Estafilocócica A/inmunología
12.
Proc Natl Acad Sci U S A ; 117(37): 22992-23000, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32855300

RESUMEN

Antibodies may bind to bacterial pathogens or their toxins to control infections, and their effector activity is mediated through the recruitment of complement component C1q or the engagement with Fcγ receptors (FcγRs). For bacterial pathogens that rely on a single toxin to cause disease, immunity correlates with toxin neutralization. Most other bacterial pathogens, including Staphylococcus aureus, secrete numerous toxins and evolved multiple mechanisms to escape opsonization and complement killing. Several vaccine candidates targeting defined surface antigens of S. aureus have failed to meet clinical endpoints. It is unclear that such failures can be solely attributed to the poor selection of antibody targets. Thus far, studies to delineate antibody-mediated uptake and killing of Gram-positive pathogens remain extremely limited. Here, we exploit 3F6-hIgG1, a human monoclonal antibody that binds and neutralizes the abundant surface-exposed Staphylococcal protein A (SpA). We find that galactosylation of 3F6-hIgG1 that favors C1q recruitment is indispensable for opsonophagocytic killing of staphylococci and for protection against bloodstream infection in animals. However, the simple removal of fucosyl residues, which results in reduced C1q binding and increased engagement with FcγR, maintains the opsonophagocytic killing and protective attributes of the antibody. We confirm these results by engineering 3F6-hIgG1 variants with biased binding toward C1q or FcγRs. While the therapeutic benefit of monoclonal antibodies against infectious disease agents may be debatable, the functional characterization of such antibodies represents a powerful tool for the development of correlates of protection that may guide future vaccine trials.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Fagocitosis/inmunología , Proteína Estafilocócica A/inmunología , Animales , Línea Celular , Glicosilación , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
13.
J Immunol Methods ; 483: 112807, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32540393

RESUMEN

Monoclonal antibodies (mAb) are unique tools in therapeutics and immunodiagnostics applications but many of these applications rely on conjugated mAbs. Whether conjugating drugs or tracers, the conjugation process, frequently taking advantage of primary amines on lysine residues, may affect the binding activity of the antibodies. Furthermore, due to the sticky nature of many mAbs, unfavorable interactions may become eminent, with the result of high background signals. The workload associated with producing mAbs, able to withstand conjugation, preserving stability and affinity and avoiding off-target interactions, is comprehensive and related with only incidental success. We designed a method, where uncloned hybridomas were pre-selected for secretion of mAbs with the above characteristics. Using human collectin K1 (CL-K1, alias CL-11, Colec11) as a model antigen, mAbs present in culture supernatant from uncloned hybridomas were immobilized on Protein A beads, followed by solid phase biotinylation and subsequent elution. ELISA was employed to compare the binding activity of conjugated vs. unconjugated mAbs, and furthermore for their application in combination with other antibodies. From a group of 96 uncloned hybridomas we accomplished in obtaining five suitable mAbs, among which, two mAbs were superior. The successful conjugation of the selected mAbs with fluorophores and subsequent applications in microscopy and flow cytometry were further demonstrated. In conclusion, pre-selection of uncloned hybridomas, by testing of their mAbs' ability to withstand conjugation with tracers or drugs, is a successful strategy to avoid a huge workload of cloning numerous hybridomas, in order to obtain conjugatable mAbs.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Colectinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inmunoconjugados/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Biotinilación , Células CHO , Clonación Molecular , Colectinas/genética , Colectinas/inmunología , Cricetulus , Humanos , Hibridomas , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Ratones , Estabilidad Proteica , Proteína Estafilocócica A/inmunología
14.
Appl Environ Microbiol ; 86(17)2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32591386

RESUMEN

Staphylococcus aureus and other coagulase-positive Staphylococcus spp. bind the Fc region of IgG antibodies through expression of protein A (SpA). These species have consequently been a source of false-positive signals in antibody-based assays designed to detect other target bacteria. Here, flow cytometry was used to study the influence of a number of factors on the SpA-mediated binding of single cells to an anti-human IgG antibody, including strain, heat killing, overnight storage, growth phase, cell physiology, surface adhesion, and growth in model food systems. Through the costaining of antibody-stained cells with the permeability dye propidium iodide and calcein violet AM, the cell physiological status was related to SpA-mediated antibody binding. Generally, permeabilized cells lacking esterase activity did not strongly bind antibody. The binding of a number of commercially available polyclonal IgG antibodies to non-Staphylococcus spp. was also characterized. Not all SpA-expressing species showed strong binding of mouse IgG, and one species not known to express SpA showed strong binding. Most SpA-expressing strains bound rabbit IgG antibodies to some extent, whereas only one strain bound goat IgG. To reduce or eliminate SpA-mediated IgG binding, the following products were evaluated as blocking reagents and applied prior to staining with primary or secondary antibody: normal rabbit serum, mouse IgG isotype control, goat IgG, and a commercial FcR blocking reagent. Only the FcR blocking reagent consistently reduced SpA-mediated binding of Staphylococcus spp. to antibodies against other species and could be recommended as a blocking reagent in immunoassays designed to detect non-Staphylococcus species.IMPORTANCE This study characterizes a widespread but little-studied problem associated with the antibody-based detection of microbes-the Staphylococcus protein A (SpA)-mediated binding of IgG antibodies-and offers a solution: the use of commercial FcR blocking reagent. A common source of false-positive signals in the detection of microbes in clinical, food, or environmental samples can be eliminated by applying this study's findings. Using flow cytometry, the authors demonstrate the extent of heterogeneity in a culture's SpA-mediated binding of antibodies and that the degree of SpA-mediated antibody binding is strain, growth phase, and food matrix dependent and influenced by simulated food processing treatments and cell adherence. In addition, our studies of SpA-mediated binding of Staphylococcus spp. to antibodies against other bacterial species produced a very nuanced picture, leading us to recommend testing against multiple strains of S. aureus and S. hyicus of all antibodies to be incorporated into any immunoassay designed to detect a non-Staphylococcus spp.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Receptores Fc/metabolismo , Proteína Estafilocócica A/inmunología , Staphylococcus/metabolismo , Anticuerpos Antibacterianos/metabolismo , Citometría de Flujo , Unión Proteica , Proteína Estafilocócica A/metabolismo
15.
BMC Res Notes ; 13(1): 135, 2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-32143691

RESUMEN

OBJECTIVES: Pythiosis is a deadly infectious disease caused by Pythium insidiosum. Reports of both human and animal pythiosis are on the rise worldwide. Prognosis of the pythiosis patients relies on early diagnosis and prompt treatment. There are needs for an immunodiagnostic test that can detect the disease in both humans and animals. This study aims at reporting an optimized protocol for the development of a protein A/G-based enzyme-linked immunosorbent assay (ELISA) for the detection of anti-P. insidiosum antibody in multiple host species. RESULTS: A total of 25 pythiosis and 50 control sera, obtained from humans, horses, dogs, cats, and cows, were recruited for the assay development. With a proper ELISA cutoff point, all pythiosis sera can ultimately be distinguished from the control sera. The successfully-developed protein A/G-based ELISA can detect the anti-P. insidiosum antibodies in serum samples of both humans and animals. It is a versatile, feasible-to-develop, and functional immunodiagnostic assay for pythiosis.


Asunto(s)
Anticuerpos/sangre , Proteínas Bacterianas/química , Ensayo de Inmunoadsorción Enzimática/métodos , Pitiosis/diagnóstico , Pythium/aislamiento & purificación , Proteína Estafilocócica A/química , Animales , Proteínas Bacterianas/inmunología , Estudios de Casos y Controles , Gatos , Bovinos , Perros , Diagnóstico Precoz , Ensayo de Inmunoadsorción Enzimática/normas , Caballos , Humanos , Sueros Inmunes/química , Pitiosis/sangre , Pitiosis/inmunología , Pitiosis/parasitología , Pythium/inmunología , Sensibilidad y Especificidad , Proteína Estafilocócica A/inmunología
16.
Sensors (Basel) ; 20(3)2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32023863

RESUMEN

The sensing properties of immunosensors are determined not only by the amount of immobilized antibodies but also by the number of effective antigen-binding sites of the immobilized antibody. Protein A (PA) exhibits a high degree of affinity with the Fc part of IgG antibody to feasibly produce oriented antibody immobilization. This work proposes a simple method to control the PA surface density on gold nanostructure (AuNS)-deposited screen-printed carbon electrodes (SPCEs) by mixing concentration-varied PA and bovine serum albumin (BSA), and to explore the effect of PA density on the affinity attachment of anti-salbutamol (SAL) antibodies by electrochemical impedance spectroscopy. A concentration of 100 µg/mL PA and 100 µg/mL BSA can obtain a saturated coverage on the 3-mercaptoproponic acid (MPA)/AuNS/SPCEs and exhibit a 50% PA density to adsorb the amount of anti-SAL, more than other concentration-varied PA/BSA-modified electrodes. Compared with the randomly immobilized anti-SAL/MPA/AuNS/SPCEs and the anti-SAL/PA(100 µg/mL):BSA(0 µg/mL)/MPA/AuNS/SPCE, the anti-SAL/PA(100 µg/mL): BSA(100 µg/mL)/MPA/AuNS/SPCE-based immunosensors have better sensing properties for SAL detection, with an extremely low detection limit of 0.2 fg/mL and high reproducibility (<2.5% relative standard deviation). The mixture of PA(100 µg/mL):BSA(100 µg/mL) for the modification of AuNS/SPCEs has great promise for forming an optimal protein layer for the oriented adsorption of IgG antibodies to construct ultrasensitive SAL immunosensors.


Asunto(s)
Albuterol/aislamiento & purificación , Técnicas Biosensibles , Inmunoensayo/métodos , Albuterol/inmunología , Anticuerpos Inmovilizados/química , Carbono/química , Oro/química , Humanos , Límite de Detección , Nanoestructuras , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/inmunología , Proteína Estafilocócica A/química , Proteína Estafilocócica A/inmunología
17.
Front Immunol ; 11: 524180, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33692774

RESUMEN

The type 1 TNF-α receptor (TNFR1) has a central role in initiating both pro-inflammatory and pro-apoptotic signaling cascades in neutrophils. Considering that TNFR1 signals Staphylococcus aureus protein A (SpA), the aim of this study was to explore the interaction of this bacterial surface protein with neutrophils and keratinocytes to underscore the signaling pathways that may determine the fate of these innate immune cells in the infected tissue during staphylococcal skin infections. Using human neutrophils cultured in vitro and isogenic staphylococcal strains expressing or not protein A, we demonstrated that SpA is a potent inducer of IL-8 in neutrophils and that the induction of this chemokine is dependent on the SpA-TNFR1 interaction and p38 activation. In addition to IL-8, protein A induced the expression of TNF-α and MIP-1α highlighting the importance of SpA in the amplification of the inflammatory response. Protein A contributed to reduce neutrophil mortality prolonging their lifespan upon the encounter with S. aureus. Signaling initiated by SpA modulated the type of neutrophil cell death in vitro and during skin and soft tissue infections (SSTI) in vivo triggering the apoptotic pathway instead of necrosis. Moreover, SpA induced pro-inflammatory cytokines in keratinocytes, modulating their survival in vitro and preventing the exacerbated necrosis and ulceration of the epithelium during SSTI in vivo. Taken together, these results highlight the importance of the inflammatory signaling induced by protein A in neutrophils and skin epithelial cells. The ability of protein A to modulate the neutrophil/epithelial cell death program in the skin is of clinical relevance considering that lysis of neutrophils and epithelial cells will promote an intense inflammatory response and contribute to tissue damage, a non-desirable feature of complicated SSTI.


Asunto(s)
Queratinocitos/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Neutrófilos/inmunología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Citocinas/inmunología , Humanos , Queratinocitos/microbiología , Neutrófilos/microbiología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
18.
J Appl Microbiol ; 128(1): 102-115, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31596989

RESUMEN

AIM: This work reports a new method for the use of lasers for the selective killing of bacteria targeted using light-absorbing Silver nanoparticles (Ag-NPs) conjugated with a specific antibody against the Gram-positive bacterium Staphylococcus aureus (S. aureus). METHODS AND RESULTS: Ag-NPs were synthesized using a chemical reduction method and characterized with respect to their surface plasmon resonance, surface morphology via transmission electron microscopy (TEM) and dynamic light scattering (DLS). The bacterial surface was targeted using 20 nm Ag-NPs conjugated with an anti-protein A antibody. Labelled bacteria were irradiated with blue visible laser at 2·04 W/cm2 . The antibacterial activity of functionalized Ag-NPs was investigated by fluorescence microscopy after irradiation, and morphological changes in S. aureus after laser treatment were assessed using scanning electron microscopy (SEM). The laser-irradiated, functionalized Ag-NPs exhibited significant bactericidal activity, and laser-induced bacterial damage was observed after 10 min of laser irradiation against S. aureus. The fluorescence microscopic analysis results supported that bacterial cell death occurred in the presence of the functionalized Ag-NPs. CONCLUSIONS: The results of this study suggest that a novel method for the preparation of functionalized nanoparticles has potential as a potent antibacterial agent for the selective killing of resistant disease-causing bacteria. SIGNIFICANCE AND IMPACT OF THE STUDY: This study shows that Ag-NPs functionalized with a specific antibody, could be used in combination with laser radiation as a novel treatment to target resistant bacterial and fungal pathogens with minimal impact on normal microflora.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Antibacterianos/química , Nanopartículas del Metal/química , Plata/química , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/química , Antibacterianos/efectos de la radiación , Rayos Láser , Pruebas de Sensibilidad Microbiana , Plata/farmacología , Especificidad de la Especie , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Propiedades de Superficie
19.
Int Immunol ; 32(2): 89-104, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-31713625

RESUMEN

Staphylococcus aureus is a main pathogen of osteomyelitis and protein A is a virulence factor with high affinity for IgG. In this study, we investigated whether S. aureus affects the differentiation and bone resorption of osteoclasts through the IgG-binding capacity of protein A. Staphylococcus aureus pre-treated with serum or IgG showed marked enhancement in osteoclastogenesis and bone resorption compared to non-treated S. aureus or a protein A-deficient mutant. Blocking of the Fc receptor and deletion of the Fcγ receptor gene in osteoclast precursor cells showed that enhanced osteoclastogenesis stimulated by S. aureus IgG immune complexes (ICs) was mediated by the Fc receptor on osteoclast precursor cells. In addition, osteoclastogenesis stimulated by S. aureus ICs but not the protein A-deficient mutant was markedly reduced in osteoclast precursor cells of Myd88-knockout mice. Moreover, NFATc1, Syk and NF-κB signals were necessary for osteoclastogenesis stimulated by S. aureus ICs. The results suggest the contribution of a of Toll-like receptor 2 (TLR2)-Myd88 signal to the activity of S. aureus ICs. We further examined the expression of pro-inflammatory cytokines that is known to be enhanced by FcγR-TLR cross-talk. Osteoclasts induced by S. aureus ICs showed higher expression of TNF-α and IL-1ß, and marked stimulation of proton secretion of osteoclasts activated by pro-inflammatory cytokines. Finally, injection of S. aureus, but not the protein A-deficient mutant, exacerbated bone loss in implantation and intra-peritoneal administration mouse models. Our results provide a novel mechanistic aspect of bone loss induced by S. aureus in which ICs and both Fc receptors and TLR pathways are involved.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Diferenciación Celular , Inmunoglobulina G/inmunología , Receptores Fc/inmunología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Receptor Toll-Like 2/inmunología , Animales , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/inmunología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Osteogénesis/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Ligando RANK/farmacología , Receptores Fc/deficiencia , Receptores Fc/genética , Proteína Estafilocócica A/genética , Staphylococcus aureus/citología , Ácidos Teicoicos/farmacología
20.
Curr Osteoporos Rep ; 17(6): 395-404, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31721069

RESUMEN

PURPOSE OF REVIEW: Staphylococcus aureus is the primary pathogen responsible for osteomyelitis, which remains a major healthcare burden. To understand its dominance, here we review the unique pathogenic mechanisms utilized by S. aureus that enable it to cause incurable osteomyelitis. RECENT FINDINGS: Using an arsenal of toxins and virulence proteins, S. aureus kills and usurps immune cells during infection, to produce non-neutralizing pathogenic antibodies that thwart adaptive immunity. S. aureus also has specific mechanisms for distinct biofilm formation on implants, necrotic bone tissue, bone marrow, and within the osteocyte lacuno-canicular networks (OLCN) of live bone. In vitro studies have also demonstrated potential for intracellular colonization of osteocytes, osteoblasts, and osteoclasts. S. aureus has evolved a multitude of virulence mechanisms to achieve life-long infection of the bone, most notably colonization of OLCN. Targeting S. aureus proteins involved in these pathways could provide new targets for antibiotics and immunotherapies.


Asunto(s)
Inmunidad Adaptativa/inmunología , Huesos/inmunología , Evasión Inmune , Osteomielitis/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/patogenicidad , Absceso/inmunología , Linfocitos B/inmunología , Biopelículas , Huesos/microbiología , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Osteoblastos/microbiología , Osteoclastos/microbiología , Osteocitos/microbiología , Osteomielitis/microbiología , Infecciones Estafilocócicas/microbiología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...