Fact-checked by Grok 2 weeks ago

Protein A

Protein A (SpA), also known as staphylococcal protein A, is a 42 kDa virulence factor and multifunctional surface protein produced by the Gram-positive bacterium Staphylococcus aureus. It is renowned for its high-affinity binding to the Fc region of immunoglobulin G (IgG) antibodies from humans and other mammals, as well as to the Fab region of certain B-cell receptors, enabling the bacterium to evade host immune responses by inhibiting phagocytosis and dysregulating B-cell activity. First identified in the 1960s and structurally characterized in the late 1970s, SpA is synthesized as a precursor with an N-terminal signal sequence for secretion, five tandem immunoglobulin-binding domains (labeled E, D, A, B, and C from N- to C-terminus), and a C-terminal region featuring an LPXTG sorting motif for anchoring to the bacterial cell wall via peptidoglycan cross-linking. In its native context, SpA contributes significantly to S. aureus pathogenesis by binding the Fcγ domain of IgG to prevent opsonization and Fc receptor-mediated uptake by immune cells such as neutrophils and macrophages, while also cross-linking VH3-family B-cell receptors to induce polyclonal B-cell expansion, apoptosis, or anergy, thereby suppressing adaptive immunity. This protein is highly conserved across S. aureus strains and can be spontaneously released into the extracellular milieu, either as a free soluble form or tethered to cell wall fragments, enhancing its immunomodulatory reach during infection. Studies have shown that SpA mutants impair bacterial virulence in animal models, underscoring its essential role in conditions ranging from skin infections to systemic diseases like sepsis. Due to its specific and robust IgG-binding properties, recombinant Protein A has revolutionized since the 1980s, serving as the in resins (e.g., Sepharose-based columns) for large-scale purification of therapeutic monoclonal antibodies, which now constitute a multibillion-dollar . Engineered derivatives, such as alkali-tolerant variants and the affibody scaffold derived from its helical binding domains, have expanded its utility in diagnostics, , and therapeutic applications, including clinical trials for cancer imaging and autoimmune treatments. Additionally, SpA-based systems have facilitated platforms, such as surface display on non-pathogenic staphylococci for development and of binding proteins.

Molecular Biology

Gene and Expression

Protein A is encoded by the spa gene, which is located on the chromosome of Staphylococcus aureus. The gene consists of approximately 1.5 kb of DNA sequence and encodes a precursor protein of 516 amino acids with a calculated molecular weight of approximately 56 kDa (mature protein ~42 kDa). The precursor includes an N-terminal signal peptide, multiple immunoglobulin-binding domains, a variable X region, a cell wall-spanning region, and a C-terminal sorting signal. Transcription of the gene is tightly regulated by global regulators in S. aureus, including the accessory gene regulator (Agr) system and the staphylococcal accessory regulator A (). The Agr system, via its effector RNAIII, represses spa expression in the post-exponential growth phase by binding to upstream promoter elements between -137 and -125 bp, resulting in up to a 14-fold reduction in transcriptional activity compared to agr-null strains. also represses spa transcription by binding to two sites in the promoter region (-97 to -91 bp and -64 to -44 bp), with the absence of SarA leading to a 40-fold increase in promoter activity for constructs including these sites. This regulation contributes to phase-variable expression of Protein A during infection, where transcription is prominent in the exponential phase for immune evasion but downregulated post-exponentially to adapt to host environments. Post-translational processing of the Protein A precursor involves cleavage of the N-terminal and anchoring to the . The is cleaved by type I signal peptidase at the YSIRK/GXXS after at position 36, facilitating secretion primarily at the septal membrane in a SecA- and lipoteichoic acid-dependent manner. Subsequently, the mature protein is anchored to the via its C-terminal LPXTG , where sortase A catalyzes cleavage between the and residues, forming an bond between the carboxyl and the amino group of a pentaglycine cross-bridge with a transpeptidation rate at least twofold faster than . The spa gene shows significant sequence variability, particularly in the X region composed of short tandem repeats, which is exploited for spa typing to identify S. aureus strains. Over 20,000 distinct spa types have been reported globally, enabling epidemiological tracking and differentiation of clones based on repeat patterns.

Structure and Domains

Protein A is a 42 kDa surface protein of Staphylococcus aureus composed of five tandemly repeated, homologous immunoglobulin G (IgG)-binding domains designated E, D, A, B, and C. While most strains encode five domains, some, such as N315, have only four. Each domain spans approximately 60 amino acids and adopts a compact left-handed three-α-helix bundle fold, with the first helix tilted relative to the antiparallel second and third helices, stabilized by a hydrophobic core. These domains are linked by short, flexible linkers, enabling independent folding while maintaining the overall linear architecture essential for multivalent interactions. The mature protein is derived from a precursor featuring an N-terminal for secretion and a C-terminal region with an LPXTG sorting motif followed by a hydrophobic domain and positively charged tail, which facilitates covalent anchoring to the bacterial peptidoglycan via sortase A-mediated transpeptidation. High-resolution , including the NMR solution of the isolated B domain (PDB ID: 1BDD) and crystal of domains in complex with antibody fragments (e.g., PDB ID: 1DEE for the D domain with IgM ), confirm the conserved helical topology and reveal surface-exposed residues critical for ligand recognition. Protein A exhibits an (pI) of approximately 5.1, reflecting its overall acidic character due to the composition. It demonstrates notable stability, retaining structural integrity across 3–11 and temperatures up to 100°C, as well as resistance to degradation by common proteases such as and under physiological conditions. These physicochemical attributes underscore its role as a durable . The IgG-binding domains display strong evolutionary conservation across S. aureus strains, with sequence identity exceeding 90% in clinical isolates, though minor polymorphisms in inter-domain linkers or surface loops can result in subtle variations in binding affinity for different IgG subclasses.

Antibody Interactions

Binding Mechanism

Protein A, a surface protein from Staphylococcus aureus, primarily binds to the Fc region of immunoglobulin G (IgG) antibodies through non-covalent interactions, including hydrogen bonds and van der Waals forces. This interaction is strongest with human IgG1, IgG2, and IgG4 subclasses, while binding to IgG3 is generally absent due to sequence variations at the interface. The five homologous IgG-binding domains (E, D, A, B, C) of Protein A each bind independently to the Fc region, with the interface primarily involving the α-helix 1 and 2 of each domain docking into a pocket at the CH2-CH3 domain junction of Fc. Key residues on Protein A, such as Phe5, Gln9, Gln10, Asn11, Phe13, Tyr14, Leu17 from helix 1, and Asn28, Ile31, Gln32, Lys35 from helix 2 (in the B domain numbering), form extensive contacts with Fc residues including Ile253, Ser254, Gln311, Leu432, and Asn434, primarily through van der Waals packing and hydrogen bonding. Upon binding, the Fc region undergoes a conformational shift, with Ile253 displacing a phenylalanine side chain in the Protein A pocket and reducing overall conformational heterogeneity at the interface, which stabilizes the complex. Binding is pH-dependent, optimal at neutral (around 7) where electrostatic attractions support the interaction, but dissociates at low (approximately 3) due to of and other residues, leading to repulsive electrostatic forces between positively charged sites on Protein A (e.g., His137, Arg146, Lys154) and . This property enables reversible binding in . Due to its five IgG-binding domains, Protein A can theoretically bind up to five IgG molecules, but steric constraints limit the observed to 1-2 IgG molecules per Protein A in solution. Protein A's modular structure facilitates multivalent interactions without inter-domain interference.

Specificity and Affinity

Protein A exhibits high affinity for the Fc region of IgG subclasses 1, 2, and 4, with constants (Kd) typically in the range of 10-100 nM, while showing no detectable binding to IgG3, IgD, or IgE. Similarly, it binds strongly to rabbit IgG with comparable nanomolar affinities, making these interactions central to its utility in immunological applications. In contrast, affinity for mouse IgG1 is notably lower, often in the micromolar range or weaker, whereas subclasses like IgG2a and IgG2b show moderate to strong binding. The five IgG-binding domains of Protein A (E, D, A, B, and C) display subtle variations in , with domain B generally exhibiting the highest strength (Kd ≈ 4-10 nM for IgG1 and IgG4) and domain E the lowest (Kd ≈ 17-50 nM for the same subclasses). These differences arise from sequence variations among the domains but do not drastically alter overall specificity, as all domains bind effectively to IgG1 and IgG4 under standard conditions. Cross-reactivity extends to other , with strong binding to IgG (similar to levels) and weak or conditional affinity for bovine IgG, which requires optimized conditions (e.g., pH 8.0) for effective interaction due to subclass-specific variations. Subclass impacts are evident across species; for instance, IgG1 binds more avidly than IgG2 in some assays, influencing elution profiles in purification. These affinities are commonly quantified using techniques such as (SPR) for kinetic parameters (association and dissociation rates yielding Kd) and (ELISA) for relative binding strengths, providing equilibrium dissociation constants that confirm Protein A's selectivity for IgG Fc over other immunoglobulin classes.
SpeciesIgG Binding Affinity to Protein ARepresentative Kd (nM) for Key Interactions
Strong (subclasses 1, 2, 4); none (3, D, E)2-15 (IgG1)
Strong~10-50
Weak (IgG1); moderate-strong (2a, 2b)>100 (IgG1); 10-50 (IgG2a)
Strong~10-100
BovineWeak/conditional>500 (requires pH optimization)

Biological Functions

Role in Pathogenesis

Protein A plays a central role in the pathogenesis of Staphylococcus aureus infections by enhancing bacterial survival through its interaction with host immunoglobulins. The protein binds to the Fc region of immunoglobulin G (IgG) antibodies on the bacterial cell wall, which sterically hinders opsonization and blocks phagocytosis by neutrophils and macrophages. This immune evasion mechanism allows S. aureus to resist clearance in the bloodstream and tissues, facilitating dissemination and persistence during infection. Beyond anti-opsonic effects, Protein A acts as a potent B-cell by engaging the region of B-cell receptors, particularly those expressing VH3 variable heavy chain domains. This binding triggers massive polyclonal B-cell proliferation and subsequent activation, culminating in widespread and depletion of the B-cell repertoire. The result is a profound suppression of , diverting the host's adaptive response and promoting bacterial proliferation without eliciting protective antibodies. In vivo, Protein A expression is upregulated during S. aureus colonization and infection, contributing directly to abscess formation and the development of chronic infections by sustaining bacterial loads in host tissues. Experimental evidence from animal models underscores its virulence potential; spa gene mutants, which lack functional Protein A, demonstrate significantly reduced pathogenicity in mouse sepsis models, with lower bacterial burdens and improved host survival compared to wild-type strains. This strain-specific effect highlights Protein A's essential contribution to the severity and outcome of S. aureus-induced disease.

Immune Evasion

Protein A contributes to Staphylococcus aureus immune evasion by synergizing with other virulence factors, such as the polysaccharide capsule and , to shield Protein A-bound IgG from host complement activation. The capsule forms a physical barrier that reduces opsonization of IgG-opsonized bacteria, while promotes clot formation around bacterial aggregates, further masking surface-bound IgG and inhibiting complement deposition and subsequent phagocytic clearance. This integrated strategy enhances bacterial survival in blood and tissues by collectively disrupting the triggered by IgG Fc regions. Protein A-IgG complexes and its superantigenic activity modulate host responses, inducing excessive production of pro-inflammatory s like TNF-α and IL-6, which can exhaust immune effectors and promote . As a B-cell , Protein A preferentially binds and activates VH3-expressing B cells, leading to polyclonal expansion and presentation of bacterial antigens to T cells, thereby triggering a that diverts adaptive immunity away from protective responses. This hyperactivation also induces shedding of TNF receptor 1, dampening sustained TNF-α signaling while amplifying acute IL-6 release, contributing to and immune dysregulation during infection. In biofilm formation and intracellular persistence, Protein A promotes multicellular aggregates by mediating cell-to-cell adhesion and production, shielding from immune detection within host tissues. Elevated Protein A expression in regulatory mutants enhances development on abiotic surfaces and during catheter-associated infections, facilitating chronic colonization. For intracellular persistence, Protein A supports survival within non-professional , such as epithelial cells, by activating TNFR1 signaling to induce inflammatory responses that aid in evading lysosomal degradation. Additionally, Protein A evades adaptive immunity by altering the B-cell repertoire through selective expansion and depletion of VH3+ B cells, creating a persistent "" in the repertoire that impairs generation of protective antibodies against other S. aureus antigens. This depletion occurs via and anergy induction, biasing humoral responses toward non-protective, Protein A-specific antibodies. Clinically, Protein A's evasion mechanisms are prominent in methicillin-resistant S. aureus (MRSA) strains, where intact Protein A expression correlates with recurrent and infections, particularly in patients. In , Protein A penetrates the compromised barrier, activating TNFR-1 on to release TSLP and IL-8, exacerbating and promoting recurrent and superinfections that resist standard therapies. This underscores Protein A's role in perpetuating chronic infections, as seen in higher MRSA persistence rates in immunocompromised hosts.

Production Techniques

Native Isolation

Native isolation of Protein A begins with the cultivation of high-producing Staphylococcus aureus strains, such as Cowan I, which is recognized for its elevated expression levels of the protein compared to other isolates like Newman. The bacteria are typically grown in nutrient broth, such as , at 37°C with shaking to promote , reaching the late stationary phase (optical density ~2-3 at 600 nm) before harvest by to yield wet cell paste. To release Protein A from the cell wall, where it is covalently anchored via its C-terminal signal to the layer, cells are subjected to . Enzymatic using lysostaphin, a zinc-dependent that specifically cleaves the pentaglycine cross-bridges in the staphylococcal , is a seminal method developed in the early ; it involves suspending the cell paste in and incubating with 100 μg/ml lysostaphin at 37°C for 2 hours, solubilizing the protein without denaturation. Mechanical disruption methods, including or glass bead milling, provide alternatives for cell breakage but may generate and shear forces that risk partial protein degradation. Historical approaches from the 1960s to also included , where cells are suspended in and heated to 80-100°C for 1-2 hours to liberate a soluble form of Protein A, leveraging its up to ~70°C while disrupting weaker wall associations. Post-lysis, the suspension is centrifuged to remove cell debris and unbroken cells, and the supernatant undergoes at 60-80% saturation to concentrate Protein A and remove soluble impurities. The precipitated proteins are against low-salt buffer and subjected to ion-exchange on DEAE-Sepharose columns, eluting Protein A with a gradient (0-0.5 M) based on its near neutrality. Final purification employs on IgG-Sepharose, where Protein A binds selectively to the Fc region of immobilized human IgG and is eluted with low-pH buffer (pH 2.8), achieving >95% purity after and lyophilization. This multi-step process, established in the 1970s, yields approximately 450-500 mg of purified Protein A from 300 g of wet cells, though challenges persist due to co-purification of other components like teichoic acids and fibronectin-binding proteins, which can reduce specificity and require additional polishing steps for biotechnological use.

Recombinant Expression

Recombinant Protein A is primarily produced using in non-pathogenic hosts to enhance safety, scalability, and control over post-translational modifications compared to native extraction from . The most common system is , where the spa gene encoding Protein A is cloned into expression vectors for high-yield production. In E. coli BL21(DE3), periplasmic secretion is achieved using the to direct the protein to the oxidizing periplasmic space, facilitating proper folding of its multiple IgG-binding domains and reducing inclusion body formation. This approach yields soluble, functional Protein A with minimal host cell protein contamination. Alternative eukaryotic systems are employed when or complex folding is desired for engineered variants. In the yeast Pichia pastoris, the spa gene is integrated into the genome under the control of the strong AOX1 promoter, enabling methanol-inducible expression and N-linked that can mimic mammalian patterns. Yields in P. pastoris can reach up to 8.8 g/L in optimized bioreactor cultures. Mammalian cell lines, such as or HEK293, may be used for expression of glycosylated Protein A variants via viral promoters like CMV, providing authentic post-translational modifications for specific therapeutic applications. Vector design involves modular spa gene fragments encoding individual IgG-binding domains (E, D, A, B, C) to produce truncated or multimeric variants. The ZZ domain, a dimer of the engineered Z domain derived from the B domain, is widely used for bivalent binding with enhanced while maintaining small size for efficient expression. Codon optimization of the sequence for the host (e.g., E. coli-preferred codons) further boosts efficiency and yield by reducing rare codon usage. Purification of recombinant Protein A typically employs a C-terminal His-tag for immobilized metal (IMAC) on Ni-NTA resins, achieving >95% purity in a single step. Optimized fermenter processes in E. coli can attain yields up to 1 g/L of culture, enabling industrial-scale production. To address cleaning challenges in bioprocessing, alkaline-stabilized variants have been developed through . For instance, the G29A mutation in the C domain confers resistance to 0.5 M NaOH, retaining >90% capacity after repeated exposure and allowing extensive column reuse without degradation. These mutants, often multimeric like , maintain native affinity for IgG while improving resin longevity. Recent advances include further engineered variants with enhanced stability for prolonged bioprocessing.

Biotechnological Applications

Industrial Purification

Protein A is widely employed in industrial-scale purification through , where it is immobilized on solid supports such as cross-linked or beads to facilitate the capture of immunoglobulins from complex mixtures like supernatants. These typically exhibit a dynamic binding capacity of approximately 20-40 mg of IgG per mL of settled , enabling efficient processing of large volumes in biopharmaceutical manufacturing. Commercial examples include MabSelect from Cytiva, which uses an alkali-stabilized recombinant Protein A on a base for robust performance, and POROS from , which feature a perfusive particle structure for high flow rates in large-scale operations. The high cost of these , often around $10,000 per liter, underscores their role as a significant expense in , though their reusability over hundreds of cycles helps mitigate this. The purification process begins with loading the feedstock at neutral (typically 7.0-8.0), where Protein A specifically binds the Fc region of antibodies under physiological conditions, as binding affinity decreases sharply at lower . This is followed by washing steps with buffers containing high salt (e.g., 0.5-1 M NaCl) to remove unbound contaminants, ensuring minimal non-specific interactions. then occurs at acidic (3.0-4.0) using buffers like 0.1 M glycine-HCl, releasing the antibodies, which are immediately neutralized to 7.0-8.0 to prevent aggregation or denaturation. A complete cycle, including loading, washing, , and regeneration, typically takes 2-4 hours, allowing for high throughput in continuous or batch modes. Regeneration protocols are essential for resin longevity, involving cleaning with 0.1-0.5 M NaOH after each cycle to remove residual proteins and lipids, with more stringent 1 M NaOH or 0.5-1 M NaCl/NaOH treatments every 10-50 cycles to sanitize and restore capacity. These steps maintain performance over 200-500 cycles, depending on feedstock quality. Compared to alternatives like ion-exchange or hydrophobic interaction chromatography, immobilized Protein A offers superior specificity, routinely achieving greater than 95% purity for monoclonal antibodies in a single step, which is critical for biopharma production where host cell proteins and aggregates must be minimized. This high selectivity reduces the need for multiple polishing steps, streamlining workflows and enhancing overall yield.

Diagnostic and Therapeutic Uses

Protein A-coated magnetic or agarose beads are widely utilized in immunoprecipitation assays to capture immunoglobulin G (IgG) antibodies and associated protein complexes from cell lysates or serum samples, enabling the study of protein-protein interactions in immunological research. These beads leverage Protein A's high affinity for the Fc region of IgG, facilitating efficient isolation without non-specific binding to other serum components. In enzyme-linked immunosorbent assay (ELISA) formats, Protein A serves as a capture reagent for quantifying IgG antibodies, particularly in diagnostic kits for detecting subclass-specific responses, such as anti-tissue transglutaminase IgG in celiac disease screening. This application enhances assay sensitivity and specificity by orienting IgG molecules for optimal antigen detection. Additionally, Protein A-conjugated beads or reagents are employed in flow cytometry to analyze B-cell populations, where they bind surface IgG on B cells to assess activation states or antigen-specific responses during immune monitoring. Affibody molecules, small engineered scaffolds derived from the helical domains of Protein A, have been developed for targeted and in cancer. These affibodies bind tumor-associated receptors such as HER2 with high and specificity, enabling () imaging. For instance, 68Ga-ABY-025 has been evaluated in clinical trials for non-invasive assessment of HER2 expression in patients, showing improved diagnostic accuracy as of 2024. Affibodies also facilitate by conjugating therapeutic payloads to tumor-specific binders, enhancing selectivity and reducing off-target effects in preclinical models. As research tools, fusion proteins combining Protein A with (GFP) enable real-time labeling and visualization of antibody-bound targets in cellular assays, such as immunoblotting or , due to the dual functionality of IgG binding and . Protein A domains have also been incorporated into formulations as adjuvants, where fusions with antigens like subunits enhance mucosal by promoting IgG-mediated uptake and immune cell activation. Protein A-based immunoadsorption columns are employed in clinical settings to treat autoimmune diseases by selectively removing pathogenic IgG autoantibodies from , as demonstrated in trials for thyroid-associated ophthalmopathy, where repeated sessions reduced antibody levels and improved clinical symptoms. In randomized controlled studies for Guillain-Barré syndrome and , these columns showed comparable efficacy to in depleting IgG, with benefits in neurological function and cardiac parameters, particularly in refractory cases. Such applications highlight Protein A's role in therapies for conditions like , where column-based removal of immune complexes alleviates disease activity without broad protein loss.

Comparative Proteins

Other Staphylococcal Binding Proteins

In addition to Protein A, Staphylococcus aureus produces several other surface-anchored binding proteins that contribute to host interaction and immune evasion, though most do not bind immunoglobulins directly. Clumping factor A (ClfA) is a microbial surface component recognizing adhesive matrix molecules (MSCRAMM) that primarily binds the γ-chain of fibrinogen, promoting bacterial clumping and adhesion to host tissues during infection. Unlike Protein A, ClfA does not interact with IgG, focusing instead on fibrin clot formation to facilitate bloodstream survival and endocarditis. Clumping factor B (ClfB), another MSCRAMM, exhibits bifunctional binding to both fibrinogen and human cytokeratin 10, the latter interaction enabling adherence to squamous epithelial cells in the nasal mucosa for colonization. ClfB's cytokeratin binding is particularly important for initial host attachment in the anterior nares, with no reported affinity for immunoglobulins. The second immunoglobulin-binding protein (Sbi) serves as a complement to Protein A by also binding the Fc region of IgG through its N-terminal domains I and II, while its C-terminal domain IV recruits complement factor C3 to inhibit opsonization. Structurally, Sbi is an elongated extracellular protein comprising four globular domains, with domains I and II facilitating IgG binding and domains III and IV linking to complement components like C3 and factor H for enhanced immune modulation. This multi-ligand capability allows Sbi to promote bacterial aggregation with IgG and disrupt complement activation, thereby aiding evasion of phagocytosis. Major histocompatibility complex analogous protein (Map) binds intercellular adhesion molecule-1 () on host cells, inhibiting leukocyte recruitment and disrupting T-cell activation, contributing to immune evasion during infection. These proteins exhibit functional overlaps with Protein A in orchestrating immune , where ClfA and ClfB support adhesion and formation, Sbi extends IgG-mediated shielding to complement interception, and Map dampens adaptive responses, collectively enhancing S. aureus .

Protein G and Protein L

Protein G is an immunoglobulin-binding protein derived from and G streptococcal , with a native molecular weight of approximately 60 kDa. It interacts with both the and regions of IgG antibodies, enabling broad binding across various subclasses. Unlike Protein A, Protein G includes a distinct albumin-binding domain that facilitates additional interactions with , enhancing its utility in certain biological contexts. Structurally, Protein G features three repeated IgG-binding domains (B1, B2, and B3) in its extracellular region, which contribute to its high-affinity binding, particularly for mouse IgG subclasses where it exhibits greater compared to Protein A. Protein L, isolated from the anaerobic bacterium Peptostreptococcus magnus, is another bacterial immunoglobulin-binding protein that specifically targets the chains of antibodies without interacting with the Fc region. This selectivity allows Protein L to bind immunoglobulins across classes (IgG, IgM, IgA, IgD, and IgE) that contain chains, making it particularly valuable for purifying -chain-dependent fragments like regions. The protein's structure includes four homologous B domains responsible for chain recognition, each comprising an alpha-helix packed against a beta-sheet scaffold. In comparison to Protein A, both Protein G and Protein L offer complementary binding profiles due to their distinct structural features: Protein G's three IgG-binding domains plus an site enable robust and interactions for whole IgG purification, while Protein L's four B domains provide specificity for kappa light chains, ideal for or isolation without interference. These differences position Protein G as a versatile alternative for broad IgG capture, especially in mouse-derived antibodies, and Protein L as a targeted tool for light chain-focused applications in .

Historical and Research Context

Discovery and Development

Protein A was first identified in 1966 by Arne Forsgren and Jan Sjöquist as an (IgG)-binding factor located on the cell wall of the Cowan I strain. This discovery stemmed from observations of pseudo-immune reactions where bacterial cells agglutinated red blood cells coated with human , revealing the specific interaction with the Fc region of IgG. The Cowan I strain was noted for its high expression of this factor, which was initially characterized through serological assays demonstrating differential binding affinities among IgG subclasses. In the early 1970s, Jan Sjöquist and colleagues advanced structural studies by purifying the protein and identifying its multi-domain architecture, consisting of tandem repeats responsible for IgG binding and a cell wall-anchoring region. These efforts included partial amino acid sequencing and electron microscopy, which highlighted the protein's modular organization and its role in bacterial immune evasion. By the 1980s, the complete amino acid sequence of the domains was determined, confirming five homologous IgG-binding units (E, D, A, B, C) followed by a hydrophobic tail and cell wall-spanning region. In 1984, Mathias Uhlen and co-workers cloned the spa gene encoding Protein A from S. aureus using a plasmid-based expression system in Escherichia coli, enabling the full genomic sequence elucidation and marking a pivotal milestone in its molecular characterization. This cloning, patented subsequently, facilitated scalable recombinant production. Early applications of Protein A emerged in the late 1970s and 1980s, particularly in immunological techniques. Fine Chemicals (now Cytiva) commercialized Protein A-Sepharose in 1975, an resin that exploited the protein's IgG-binding properties for efficient purification of antibodies from complex mixtures, revolutionizing bioprocessing workflows. By the 1980s, radiolabeled Protein A was widely adopted in radioimmunoassays to quantify antigens and antibodies, offering a sensitive alternative to traditional methods due to its specific Fc-binding without interfering with antigen-antibody interactions. This tool facilitated advancements in diagnostics and research, such as detecting immune complexes in . The evolution toward recombinant production addressed limitations of native Protein A isolation, which relied on laborious extraction from bacterial cultures and yielded variable purity. In the mid-1980s, following gene cloning, Uhlen's team achieved the first recombinant expression of functional Protein A in E. coli, producing high yields of the protein and its domains for consistent supply. This breakthrough enabled scalable manufacturing and customization, such as truncation variants for enhanced stability in applications, laying the foundation for Protein A's widespread biotechnological use.

Recent Advances

In the 2020s, structural analyses of Protein A-IgG interactions have advanced through high-resolution techniques, revealing detailed binding interfaces that inform efforts. For instance, in the Fc region of mouse IgG2a and IgG2b, such as those enhancing Protein A by up to 8-fold, have been developed to facilitate selective purification of bispecific antibodies by exploiting differential binding strengths during . These modifications allow one arm of the bispecific construct to bind strongly while the other is weakened, enabling single-step isolation without disrupting overall antibody functionality. Recent pathogenesis research from 2022 to 2024 has highlighted the role of gene polymorphisms in driving (MRSA) evolution and persistence. Studies of long-term MRSA carriers revealed within-host mutation rates of approximately 4.9 single-nucleotide polymorphisms per genome per year, with spa typing showing diverse genotypes that contribute to strain adaptation and transmission across populations. Additionally, Protein A has been implicated in failure through induction of IL-10, an immunosuppressive that promotes antibody sialylation and impairs protective responses; blocking IL-10 during immunization restored vaccine efficacy in murine models of S. aureus . This mechanism underscores Protein A's contribution to immune evasion, as it triggers regulatory T cells that secrete IL-10, suppressing IgG-mediated immunity. Biotechnological innovations include engineered Protein A variants optimized for continuous chromatography processes. A 2020 study introduced an improved Protein A resin for countercurrent tangential , achieving higher productivity and steady-state operation compared to batch methods, with potential extensions in recent patents for enhanced stability under continuous flow. To address cost barriers—where traditional Protein A resins exceed $8,000 per liter—mimetics such as polymers and nanoCLAMP scaffolds have emerged as alkaline-stable, potentially lower-cost alternatives that maintain high IgG specificity and reusability. Emerging 2025 research utilizing single-cell sequencing has uncovered heterogeneity in S. aureus during bacteremia, including variable transcription levels across individual cells that influence and host-pathogen dynamics. Furthermore, detoxified Protein A show promise as components in S. aureus vaccines; a 2025 bivalent SpA-LukAB formulation, combined with TH1 adjuvants, elicited neutralizing antibodies and reduced bacterial burden in animal models, suggesting its potential to counter immune evasion when integrated into multi-antigen strategies.

References

  1. [1]
    The many virtues of staphylococcal protein A: A journey from N to C ...
    This review outlines the historical development and versatile applications of one of the most well-studied bacterial proteins, namely the immunoglobulin ...3. Exploiting Ig-Binding... · 3.4. Affibody Molecules And... · 4. The Cell Wall Anchoring...
  2. [2]
    Natural Human Immunity Against Staphylococcal Protein A Relies ...
    Mar 10, 2022 · Staphylococcal protein A (SpA) is a multifunctional, highly conserved virulence factor of Staphylococcus aureus. By binding the Fc portion ...
  3. [3]
    Staphylococcus Protein A - an overview | ScienceDirect Topics
    SpA is an immunoglobulin (Ig)-binding protein that exists on the bacterial surface and is spontaneously secreted into the extracellular environment.
  4. [4]
    Protein A Is Released into the Staphylococcus aureus Culture ... - NIH
    The immunoglobulin binding protein A (SpA) of Staphylococcus aureus is synthesized as a precursor with a C-terminal sorting signal.
  5. [5]
    Role of Protein A in the Evasion of Host Adaptive Immune ...
    Aug 27, 2013 · Protein A (SpA), a surface molecule of S. aureus, binds the Fcγ domain of immunoglobulin (Ig) and cross-links the Fab domain of VH3-type B cell ...
  6. [6]
    Release of protein A from the cell wall of Staphylococcus aureus
    We demonstrate that protein A of Staphylococcus aureus, a B cell superantigen, is released with peptidoglycan linked to its C terminus.
  7. [7]
  8. [8]
    Suppression of conformational heterogeneity at a protein ... - NIH
    Staphylococcal protein A (SpA) is an important virulence factor found on the surface of S. aureus cells. This 42-kDa protein has two functional halves: the N- ...
  9. [9]
    Multiscale Conformational Heterogeneity in Staphylococcal Protein A
    Oct 7, 2014 · Crystal structure of a Staphylococcus aureus protein A domain complexed with the Fab fragment of a human IgM antibody: structural basis for ...
  10. [10]
    1BDD: STAPHYLOCOCCUS AUREUS PROTEIN A ... - RCSB PDB
    The three-dimensional solution structure of the recombinant B domain (FB) of staphylococcal protein A, which specifically binds to the Fc portion of ...Missing: IgG | Show results with:IgG
  11. [11]
    P02976 · SPA_STAA8 - UniProt
    Cell wall anchoring is conferred by the LPXTG motif and following sequences (residues 482-516) which are necessary and sufficient to target other proteins to ...<|separator|>
  12. [12]
    Release of protein A from the cell wall of Staphylococcus aureus - NIH
    Cell wall-anchored surface proteins are synthesized as precursors with N-terminal signal peptides and C-terminal LPXTG motif sorting signals (9). Following ...
  13. [13]
    Crystal structure of a Staphylococcus aureus protein A domain ...
    The common bacterial pathogen, Staphylococcus aureus, produces a 42-kDa factor, protein A (SpA), that contains five highly homologous extracellular Ig-binding ...
  14. [14]
    Suppression of conformational heterogeneity at a protein ... - PNAS
    Red residues are identical in all five SpA protein binding domains. Although the Fc molecules from the new complex and from 1FC2 adopt very similar ...
  15. [15]
  16. [16]
    Study of Binding between Protein A and Immunoglobulin G Using a ...
    It is known that Protein A has high affinity for the Fc portion of IgG (Boyle and Reis, 1987). The binding between Protein A and IgG leads to formation of ...
  17. [17]
    Protein A/G - an overview | ScienceDirect Topics
    Protein G binds all human subclasses (Kd ~2×10−10 M),104,105 whereas protein A generally only binds IgG1, IgG2, and IgG4 (Kd ~2×10−9 M), but not IgG3. Because ...
  18. [18]
  19. [19]
    Antibody Binding Affinities to Protein A & Protein G | Bio-Rad
    Antibodies have different binding affinities to protein A and protein G. These properties are especially critical when deciding whether to use protein A or ...
  20. [20]
    Protein A/G Affinity | Bio-Rad
    Protein A and G chromatography media are used in antibody purification due to the high binding affinity and specificity with the antibody Fc region.
  21. [21]
    [PDF] Affinity Determination of Protein A Domains to IgG subclasses by ...
    Feb 22, 2008 · It is known that protein A bind specifically to the IgG subclasses. IgG1, IgG2, and IgG4, but lacks detectable binding to the subclass IgG3.
  22. [22]
    Comparison of Antibody IgG Binding Proteins
    Protein A is generally preferred for rabbit, pig, dog and cat IgG. Protein G has better binding capacity for a broader range of mouse and human IgG subclasses ( ...
  23. [23]
    [PDF] Protein-A-G -Affinity-for-IgG-subclasses - AMSBIO
    Hamster IgG. +. ++. ++. Strong affinity. Guinea Pig IgG. ++. +. +. Moderate/slight affinity. Bovine IgG. +. ++. +/–. Requires evaluation. Horse IgG. +. ++. -.
  24. [24]
    Conditions for binding bovine IgG1 to protein A-sepharose
    Conditions were established so that both subclasses of bovine IgG were bound to Protein A-Sepharose. Increasing the pH of the starting buffer to pH 8.0 from ...<|control11|><|separator|>
  25. [25]
    The immune evasion roles of Staphylococcus aureus protein A ... - NIH
    These proteins play a key role in inhibiting development of a long-term protective immune response by mechanisms ranging from dysregulation of the complement ...
  26. [26]
    Staphylococcus aureus infection induces protein A–mediated ... - NIH
    S. aureus expresses the virulence factor Protein A (SpA) on all clinical isolates, and SpA has been shown in mice to expand and ablate variable heavy 3 ( ...Missing: upregulated | Show results with:upregulated
  27. [27]
    Staphylococcus aureus Protein A Promotes Immune Suppression
    Oct 1, 2013 · Indeed, S. aureus produces numerous molecules that can potentially promote immune evasion, including protein A (SpA), an immunoglobulin (Ig)- ...
  28. [28]
    Mouse models for infectious diseases caused by Staphylococcus ...
    Mutants that display the most severe defects harbor mutations in genes for the following surface proteins: staphylococcal protein A (SpA), iron-regulated ...
  29. [29]
    Staphylococcal manipulation of host immune responses - Nature
    Aug 14, 2015 · To achieve this, S. aureus deploys an arsenal of immune-evasive strategies that together prevent phagocytosis and killing by neutrophils.
  30. [30]
    Antibodies against a secreted product of Staphylococcus aureus ...
    Feb 15, 2016 · S. aureus secretes coagulase (Coa), which activates host prothrombin and generates fibrin fibrils that protect the pathogen against phagocytosis ...
  31. [31]
    The immune evasion roles of Staphylococcus aureus protein A and ...
    In this review we describe the recent progress made in dissecting S. aureus immune evasion, focusing on the role of the superantigen, staphylococcal protein A ...
  32. [32]
    Shedding of Tumor Necrosis Factor Receptor 1 Induced by Protein A ...
    S. aureus protein A plays a key role in regulating inflammation through its ability to bind and signal through the TNF-α receptor 1 (TNFR1). In ...
  33. [33]
    Protein A-Mediated Multicellular Behavior in Staphylococcus aureus
    Extracellular protein A induces biofilm formation. Protein A belongs to the LPXTG family of proteins characterized by its covalent linkage to the cell wall ...
  34. [34]
    A B Cell Superantigen–Induced Persistent “Hole” in the B-1 Repertoire
    The bacterial toxin protein A from Staphylococcus aureus (SpA) interacts with B cell antigen receptors encoded by variable region heavy chain (VH) clan III.
  35. [35]
    Interactions Between Atopic Dermatitis and Staphylococcus aureus ...
    Staphylococcus aureus commonly colonizes the skin of atopic dermatitis (AD) patients and contributes to the development and exacerbation of AD.
  36. [36]
    Potential Therapeutic Targets for Combination Antibody Therapy ...
    Other virulence factors, such as coagulases (Coa) and staphylococcal protein A (SpA), aid in immune evasion by blocking phagocytosis or activating host zymogens ...
  37. [37]
    Molecular basis of surface anchored protein A deficiency in the ...
    Aug 31, 2017 · The S. aureus isolate Wood 46 has played an important role in studies designed to examine the biological function of protein A and its role in ...
  38. [38]
    [PDF] Molecular basis of surface anchored protein A deficiency in the ...
    Aug 31, 2017 · In contrast, Cowan 1 and the. Newman WT isolate, both reported to be high producers of Protein A bound 69% and 83% respectively (Fig 1), ...
  39. [39]
  40. [40]
    Novel signal peptides improve the secretion of recombinant ...
    May 12, 2017 · In order to improve secretion of recombinant proteins in E. coli ... protein A expression in Escherichia coli BL21 (DE3). Biotechnol Appl ...<|control11|><|separator|>
  41. [41]
    Secretion of recombinant proteins from E. coli - PMC - PubMed Central
    Recombinant protein expression and localization in the cytoplasm results in a high accumulation of proteins that are at risk of degradation by proteases and are ...
  42. [42]
    Pichia pastoris: A highly successful expression system for optimal ...
    Another advantage of P. pastoris as a protein production host is its ability to perform posttranslational modifications such as O‐ and N‐linked glycosylation ...
  43. [43]
    Overview of Protein Expression by Mammalian Cells - PMC
    In recent years, mammalian cells have been used in the production of recombinant proteins, antibodies, viruses, viral‐subunit proteins, and gene‐therapy vectors ...
  44. [44]
    Improved Performance of Recombinant Protein A Immobilized on ...
    Apr 28, 2017 · In this work, we report the successful site-specific covalent conjugation for recombinant protein A (ZZZ protein, three tandem Z domains) ...<|separator|>
  45. [45]
    Codon Optimization for Increased Protein Expression | GenScript
    Codon optimization uses synonymous mutations to favor efficient protein expression, addressing codon bias and rare codons that can reduce expression.Missing: spa ZZ domain dimer
  46. [46]
    His-tagged Proteins–Production and Purification - ES
    Expressed His-tagged proteins can be purified and detected easily because the string of histidine residues binds to several types of immobilized metal ions.The Histidine Tag · Protein Preparation Handbook · Nickel, Cobalt And Copper
  47. [47]
    Practical protocols for production of very high yields of recombinant ...
    Increasing the glucose concentration increases the culture cell density and protein yield. With 1.0% glucose, the OD600 reached to 7.4 and protein yield seems ...
  48. [48]
    Remarkable alkaline stability of an engineered protein A as ...
    Jul 19, 2013 · Protein A affinity chromatography is the standard purification process for the capture of therapeutic antibodies. The individual IgG-binding ...Results · Biacore Analysis Of Alkaline... · Biacore Analysis On Alkaline...
  49. [49]
    Investigation of alkaline effects on Protein A affinity ligands and ...
    Jan 1, 2021 · In this study, the enhanced alkaline stability of Protein A ligands and resins designed by protein engineering approaches is demonstrated.Missing: reuse | Show results with:reuse
  50. [50]
    Purification Using Protein A-based Chromatography Media
    ### Summary of Protein A Chromatography
  51. [51]
    [PDF] HiTrap™ rProtein A FF HiTrap Protein A HP HiTrap Protein G HP
    As a reference, the total binding capacity for human IgG is approximately 20 mg IgG/mL resin for HiTrap Protein A HP and approximately 25 mg IgG/mL resin ...
  52. [52]
    Affinity chromatography for antibody protein purification - Cytiva
    Affinity chromatography separates proteins based on a reversible binding interaction between the target protein and a ligand.Missing: industrial | Show results with:industrial
  53. [53]
    Dynamic Control to Maximize the Performance of Protein A Resin in ...
    Mar 28, 2025 · A typical Protein A resin costs around $15K per liter as of 201915. ... Since the cost of the Protein A resin can be as high as 30% of the ...
  54. [54]
    Everything You Need to Know About Protein A Affinity Resins
    Jul 10, 2025 · Use 0.1–0.5 M NaOH as a cleaning solution if the resin is alkali ... Circulate NaOH through the column for 15–30 minutes after each use.
  55. [55]
    [PDF] 1 Protein A CIP Resin Cat. No. L00433 Technical ... - GenScript
    A commonly adopted CIP protocol is to use 0.1 M. NaOH every cycle and 0.5 M NaOH every 10 cycles. VI. Storage. Store the regenerated Protein A CIP Resin in ...
  56. [56]
    Comparing Performance of New Protein A Resins for Monoclonal ...
    Feb 16, 2018 · A typical Protein A chromatography step yields >99% pure mAb, (i.e. >99% free of process impurities such as growth media components, host cell ...<|control11|><|separator|>
  57. [57]
    Protein A Chromatography Resins | Thermo Fisher Scientific - US
    A high-performance Protein A chromatography resin, specifically designed to help improve the productivity and efficiency of your antibody purification process.
  58. [58]
    Multiplex IP-FCM (immunoprecipitation-flow cytometry)
    Immunoprecipitation detected by flow cytometry (IP-FCM) is a robust, biochemical method that can be used for measuring physiologic protein-protein interactions ...
  59. [59]
    Immunoprecipitation with Dynabeads Protein A - US
    Immunomagnetic protein isolation using Dynabeads® Protein A provides a fast and reliable method for capturing Ig for small scale purification or downstream ...
  60. [60]
    Protein A and protein G ELISA for the detection of IgG ...
    Fig. 3. Median tissue transglutaminase (tTG) antibody levels detected with ELISA using protein A (PA-tTG), protein G (PG-tTG), anti-human IgG (IgG-tTG), and ...
  61. [61]
    Overview of ELISA | Thermo Fisher Scientific - US
    Plates pre-coated with Protein A, G, or A/G can help orient capture antibodies properly and preserve their antigen binding capability. Fusion proteins can be ...
  62. [62]
    Protein A-Mediated Binding of Staphylococcus spp. to Antibodies in ...
    Protein A-Mediated Binding of Staphylococcus spp. to Antibodies in Flow Cytometric Assays and Reduction of This Binding by Using Fc Receptor Blocking Reagent.Results · Purified Mouse Igg Binds To... · Flow Cytometry
  63. [63]
    Improved tumor imaging and therapy via i.v. IgG–mediated time ...
    High-dose IgG administration enhances tumor contrast in immuno-PET imaging. With the foregoing evidence that blood clearance of radiolabeled antibodies was ...Results · Fcrn Blockade Enhances The... · Figure 1. Fcrn Blockade...
  64. [64]
    Protein–Antibody Conjugates (PACs) – A Plug-and-Play Strategy for ...
    We report here on protein-antibody conjugates (PACs) that are used for antibody-directed delivery of protein therapeutics to specific cells.
  65. [65]
    Extended plasma half-life of albumin-binding domain fused human ...
    Mar 10, 2021 · Half-life extension using serum albumin-binding DARPin® domains. ... The use of gene fusions to protein A and protein G in immunology and ...
  66. [66]
    [23] Application of green fluorescent protein-protein a fusion protein ...
    This chapter discusses the construction of a protein A-GFP fusion (PA-GFP) and its use as a labeled antibody-specific ligand in immunoblotting.
  67. [67]
    Advances in vaccine adjuvants | Nature Biotechnology
    They created a gene fusion of the A1 subunit of CT and the immunoglobulin binding domain of protein A from Staphylococcus aureus. The construct was shown to be ...
  68. [68]
    Application of protein A immunoadsorption in thyroid-associated ...
    Jul 20, 2025 · This study plans to provide effective clinical evidence for protein A immunoadsorption treatment of TAO by comparing the efficacy and safety ...
  69. [69]
    Study Details | NCT05114941 | Comparison of the Efficacy and ...
    This study aims to explore the efficacy and safety of protein A immunoadsorption and intravenous immunoglobulin in the treatment of Guillain-Barre syndrome.
  70. [70]
    Immunoadsorption in SLE: Three different high affinity columns are ...
    Dec 29, 2009 · The American College of Rheumatology response criteria for systemic lupus erythematosus clinical trials: Measures of overall disease activity.
  71. [71]
    Immune Evasion by Staphylococcus aureus | Microbiology Spectrum
    The first evasion molecule described to have antiopsonic properties is staphylococcal protein A (SpA). SpA is linked to the staphylococcal cell wall via its ...
  72. [72]
    Clumping factor B (ClfB), a new surface-located fibrinogen ... - PubMed
    The surface-located fibrinogen-binding protein (clumping factor; ClfA) of Staphylococcus aureus has an unusual dipeptide repeat linking the ligand binding ...
  73. [73]
    Identification of the Staphylococcus aureus MSCRAMM clumping ...
    Clumping factor B (ClfB) of Staphylococcus aureus binds to cytokeratin 10 and to fibrinogen. In this study the binding site in human fibrinogen was localized to ...
  74. [74]
    Structure-Function Analysis of the C3 Binding Region of ...
    Sbi domains I and II (Sbi-I and Sbi-II) bind IgG. Sbi domain IV (residues 198–266) binds the central complement protein C3. When linked to Sbi-III, Sbi-IV ...
  75. [75]
    S. aureus IgG-binding proteins SpA and Sbi - ScienceDirect.com
    The putative extracellular region of Sbi (Sbi-E) is an elongated molecule consisting of four major globular domains (I, II, III and IV) (Burman et al., 2007).
  76. [76]
    The Staphylococcus aureus Protein Sbi Acts as a Complement ...
    Sbi is a multifunctional bacterial protein, which binds host complement components Factor H and C3 as well as IgG and β 2 -glycoprotein I and interferes with ...
  77. [77]
    The Staphylococcus aureus Map protein is an immunomodulator ...
    These data suggest a role for Map as an immunomodulatory protein that may play a role in persistent SA infections by affecting protective cellular immunity.
  78. [78]
    Protein G genes: structure and distribution of IgG-binding ... - PubMed
    Protein G has also been shown to bind human serum albumin but at a site that is structurally separated from the IgG-binding region. From the known gene sequence ...
  79. [79]
    Streptococcal protein G has affinity for both Fab- and Fc-fragments of ...
    We have analyzed the binding of IgG and fragments of IgG [Fc, F(ab')2, and Fab] to group C and G streptococci and to protein G, the IgG binding cell wall ...
  80. [80]
    and albumin-binding regions of streptococcal protein G.
    Protein G, the immunoglobin G-binding surface protein of group C and G streptococci, also binds serum albumin.
  81. [81]
    Crystal structure of the C2 fragment of streptococcal protein G in ...
    The crystal structure of a single Ig-binding domain of streptococcal protein G was determined at 3.5 å resolution in complex with the Fc fragment of human IgG.
  82. [82]
    Protein A and Protein G Purification of Antibodies - PubMed
    Jan 2, 2019 · IgGs from most species bind to Protein G at near physiological pH and ionic strength with a higher affinity than IgG binding to Protein A.
  83. [83]
    Protein L from Peptostreptococcus magnus binds to the kappa light ...
    Extensive reduction and alkylation of the V kappa fragment or the native kappa chain resulted in complete loss of protein L binding. Although it is possible, ...
  84. [84]
    Purification of antibodies using protein L-binding framework ...
    Protein L from the bacterial species Peptostreptococcus magnus binds specifically to the variable domain of Ig light chains, without interfering with the ...
  85. [85]
    Structure of peptostreptococcal protein L and identification ... - PubMed
    An Escherichia coli high level expression system was adapted for the production of large amounts of two Ig-binding protein L fragments comprising one and four B ...
  86. [86]
    Recombinant Protein L: Production, Purification and ...
    Nov 20, 2022 · Protein L (PpL) is a universal binding ligand that can be used for the detection and purification of antibodies and antibody fragments.
  87. [87]
    Antiphagocytic effects of staphylococcal protein A - PubMed
    Antiphagocytic effects of staphylococcal protein A. J Immunol. 1969 Dec;103(6):1405-10. Authors. J H Dossett, G Kronvall, R C Williams Jr, P G Quie. PMID ...Missing: discovery 1966 original paper
  88. [88]
    Process Chromatography: Five Decades of Innovation
    Feb 2, 2007 · Throughout this period, Pharmacia Fine Chemicals dominated the chromatographic separations industry, launching Sepharose in 1966, Protein A ...
  89. [89]
    [PDF] Biopharmaceutical Manufacturing: Historical Perspectives and ...
    Pharmacia Fine Chemicals released the first commercialized Protein A resin, Protein A Sepharose, in 1975, and its ability to purify immunoglobulins from ...
  90. [90]
    [PDF] protein A gene fusion vectors - EMBO Press
    In contrast, S. aureus is probably better for expressing truncated protein A molecules (Uhlen et al., 1984c) or fusion proteins consisting of protein A and ...
  91. [91]
    Modulation of protein A binding allows single-step purification ... - NIH
    IgG binds Z-domain using three loops in the Fc region, two from the CH2 and one from CH3 domain. By sequence comparison of mouse and human IgG Fc regions ...
  92. [92]
    Rapid Purification of Human Bispecific Antibodies via Selective ...
    Nov 14, 2017 · One advantage of this mutation set is that the total disruption in protein A binding caused by the mutations may allow larger pH differences in ...Results · Fcrn Interaction And... · Protein A Affinity...
  93. [93]
    Within-host genomic evolution of methicillin-resistant ...
    Jan 11, 2024 · This study aims to describe the within-host evolution in 121 long-term MRSA carriers with a carriage duration of 4 to 11 years by analyzing genome data on 424 ...
  94. [94]
    Interleukin 10 drives Staphylococcus aureus imprinting and vaccine ...
    Dec 16, 2024 · We show that S. aureus induces IL-10 which abrogates protective anti-staphylococcal antibody function through hyper-sialylation.Pathogenesis versus... · IL-10 drives vaccine failure in... · Limitations and outlook
  95. [95]
    Staphylococcus aureus Protein A Induces Human Regulatory T ...
    Sep 30, 2020 · These cells secrete IL-10, a cytokine associated with suppression of antigen presentation and T cell responses (31, 32). Another important cell ...
  96. [96]
    Improved protein A resin for antibody capture in a continuous ...
    Continuous countercurrent tangential chromatography (CCTC) enables steady-state continuous bioprocessing with low-pressure operation and high productivity.<|separator|>
  97. [97]
    Protein engineering of a nanoCLAMP antibody mimetic scaffold as a ...
    Jun 12, 2023 · This work describes a protein engineering campaign to develop a more robust nanoCLAMP scaffold compatible with harsh bioprocessing conditions.
  98. [98]
    IgG-Binding Peptidomimetic Mixed-Charge Polymer-Modified ...
    Nov 26, 2024 · The column packing material modified with protein A mimetic polymers was shown to be able to switch the retention and elution behavior of ...
  99. [99]
    Staphylococcus aureus transcriptomics and single-cell sequencing ...
    Sep 15, 2025 · Spa typing, which is based on the DNA sequence of the variable number tandem repeat region in the gene encoding protein A (spa), is unique in ...
  100. [100]
    A SpA+LukAB vaccine targeting Staphylococcus aureus evasion ...
    Apr 20, 2025 · This study introduces a novel S. aureus vaccine strategy by targeting immune evasion factors SpA and LukAB, utilizing potent TH1 adjuvants, and ...