Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 919
Filtrar
1.
J Phys Chem B ; 128(33): 7987-7996, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39115184

RESUMEN

Mutational changes that affect the binding of the C2 fragment of Streptococcal protein G (GB1) to the Fc domain of human IgG (IgG-Fc) have been extensively studied using deep mutational scanning (DMS), and the binding affinity of all single mutations has been measured experimentally in the literature. To investigate the underlying molecular basis, we perform in silico mutational scanning for all possible single mutations, along with 2 µs-long molecular dynamics (WT-MD) of the wild-type (WT) GB1 in both unbound and IgG-Fc bound forms. We compute the hydrogen bonds between GB1 and IgG-Fc in WT-MD to identify the dominant hydrogen bonds for binding, which we then assess in conformations produced by Mutation and Minimization (MuMi) to explain the fitness landscape of GB1 and IgG-Fc binding. Furthermore, we analyze MuMi and WT-MD to investigate the dynamics of binding, focusing on the relative solvent accessibility of residues and the probability of residues being located at the binding interface. With these analyses, we explain the interactions between GB1 and IgG-Fc and display the structural features of binding. In sum, our findings highlight the potential of MuMi as a reliable and computationally efficient tool for predicting protein fitness landscapes, offering significant advantages over traditional methods. The methodologies and results presented in this study pave the way for improved predictive accuracy in protein stability and interaction studies, which are crucial for advancements in drug design and synthetic biology.


Asunto(s)
Proteínas Bacterianas , Enlace de Hidrógeno , Inmunoglobulina G , Simulación de Dinámica Molecular , Mutación , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Inmunoglobulina G/genética , Unión Proteica , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo
2.
MAbs ; 16(1): 2379560, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39028186

RESUMEN

The self-association of therapeutic antibodies can result in elevated viscosity and create problems in manufacturing and formulation, as well as limit delivery by subcutaneous injection. The high concentration viscosity of some antibodies has been reduced by variable domain mutations or by the addition of formulation excipients. In contrast, the impact of Fc mutations on antibody viscosity has been minimally explored. Here, we studied the effect of a panel of common and clinically validated Fc mutations on the viscosity of two closely related humanized IgG1, κ antibodies, omalizumab (anti-IgE) and trastuzumab (anti-HER2). Data presented here suggest that both Fab-Fab and Fab-Fc interactions contribute to the high viscosity of omalizumab, in a four-contact model of self-association. Most strikingly, the high viscosity of omalizumab (176 cP) was reduced 10.7- and 2.2-fold by Fc modifications for half-life extension (M252Y:S254T:T256E) and aglycosylation (N297G), respectively. Related single mutations (S254T and T256E) each reduced the viscosity of omalizumab by ~6-fold. An alternative half-life extension Fc mutant (M428L:N434S) had the opposite effect in increasing the viscosity of omalizumab by 1.5-fold. The low viscosity of trastuzumab (8.6 cP) was unchanged or increased by ≤2-fold by the different Fc variants. Molecular dynamics simulations provided mechanistic insight into the impact of Fc mutations in modulating electrostatic and hydrophobic surface properties as well as conformational stability of the Fc. This study demonstrates that high viscosity of some IgG1 antibodies can be mitigated by Fc mutations, and thereby offers an additional tool to help design future antibody therapeutics potentially suitable for subcutaneous delivery.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Mutación , Omalizumab , Trastuzumab , Humanos , Trastuzumab/química , Viscosidad , Omalizumab/química , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Inmunoglobulina G/química , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética
3.
J Virol ; 98(7): e0070724, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38953655

RESUMEN

Human norovirus was discovered more than five decades ago and is a widespread cause of outbreaks of acute gastroenteritis. There are no approved vaccines or antivirals currently available. However, norovirus inhibitors, including capsid-specific monoclonal antibodies (Mabs) and nanobodies, have recently shown promising results. Several Mabs and nanobodies were found to inhibit norovirus replication using a human intestinal enteroid (HIE) culture system and/or could block norovirus attachment to histo-blood group antigen (HBGA) co-factors. In our pursuit to develop a single broad-spectrum norovirus therapeutic, we continued our analysis and development of a cross-reactive and HBGA interfering nanobody (NB26). To improve NB26 binding capacity and therapeutic potential, we conjugated NB26 onto a human IgG Fc domain (Fc-NB26). We confirmed that Fc-NB26 cross-reacts with genetically diverse GII genotype capsid protruding (P) domains (GII.8, GII.14, GII.17, GII.24, GII.26, and GII.NA1) using a direct enzyme-linked immunosorbent assay. Furthermore, X-ray crystallography structures of these P domains and structures of other GII genotypes reveal that the NB26 binding site is largely conserved, validating its broad reactivity. We showed that Fc-NB26 has ~100-fold higher affinity toward the norovirus P domain compared to native NB26. We also found that both NB26 and Fc-NB26 neutralize human norovirus replication in the HIE culture system. Furthermore, the mode of inhibition confirmed that like NB26, Fc-NB26 caused norovirus particle disassembly and aggregation. Overall, these new findings demonstrate that structural modifications to nanobodies can improve their therapeutic potential.IMPORTANCEDeveloping vaccines and antivirals against norovirus remains a challenge, mainly due to the constant genetic and antigenic evolution. Moreover, re-infection with genetically related and/or antigenic variants is not uncommon. We further developed our leading norovirus nanobody (NB26) that indirectly interfered with norovirus binding to HBGAs, by converting NB26 into a dimeric Fc-linked Nanobody (Fc-NB26). We found that Fc-NB26 had improved binding affinity and neutralization capacity compared with native NB26. Using X-ray crystallography, we showed this nanobody engaged highly conserved capsid residues among genetically diverse noroviruses. Development of such broadly reactive potent therapeutic nanobodies delivered as a slow-releasing prophylactic could be of exceptional value for norovirus outbreaks, especially for the prevention or treatment of severe acute gastroenteritis in high-risk groups such as the young, elderly, and immunocompromised.


Asunto(s)
Infecciones por Caliciviridae , Proteínas de la Cápside , Norovirus , Anticuerpos de Dominio Único , Norovirus/genética , Norovirus/efectos de los fármacos , Norovirus/inmunología , Humanos , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/química , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Infecciones por Caliciviridae/inmunología , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/terapia , Antivirales/farmacología , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/química , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas , Cápside/metabolismo , Cápside/inmunología , Antígenos de Grupos Sanguíneos/metabolismo , Replicación Viral/efectos de los fármacos , Gastroenteritis/virología , Inmunoglobulina G/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología
4.
Int J Biol Macromol ; 276(Pt 2): 133962, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39029833

RESUMEN

Covalent and oriented immobilization of antibodies (Abs) can substantially improve the sensitivity and stability of solid-phase immunoassays. By modifying the natural Abs with functional groups that provide unique handles for further conjugation, Abs could be immobilized onto the solid matrices with uniform orientation. Herein, an effective approach for Fc-specific modification of Abs was developed for the oriented and covalent immobilization of Abs. Twelve photoreactive Z-domain variants, incorporated with a photoactivable probe (p-benzoyl-L-phenylalanine, Bpa) at different positions and carrying a C-terminal Cys-tag (i.e. ZBpa-Cys variants), were individually constructed and produced in Escherichia coli and tested for photo-cross-linking to various IgGs. The different ZBpa-Cys variants demonstrated large differences in photo-conjugation efficiency for the tested IgGs. The conjugation efficiencies of 17thZBpa-Cys ranged from 90 % to nearly 100 % for rabbit IgG and mouse IgG2a, IgG2b and IgG3. Other variants, including 5thZBpa-Cys, 18thZBpa-Cys, 32thZBpa-Cys, and 35thZBpa-Cys, also displayed conjugation efficiencies of 61 %-83 % for mouse IgG1, IgG2a and IgG3. Subsequently, the photo-modified Abs, namely IgG-Cys conjugates, were covalently immobilized onto a maleimide group-functionalized solid-phase carrier on the basis of the reaction of sulfhydryl and maleimide. Thus, a generic platform for the controlled and oriented immobilization of Abs was developed, and the efficacy and potential of the proposed approach for sensitive immunoassays was demonstrated by detecting human α-fetoprotein.


Asunto(s)
Anticuerpos Inmovilizados , Cisteína , Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Cisteína/química , Animales , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Fragmentos Fc de Inmunoglobulinas/química , Anticuerpos Inmovilizados/química , Anticuerpos Inmovilizados/inmunología , Ratones , Conejos , Fenilalanina/química , Fenilalanina/análogos & derivados , Inmunoensayo/métodos , Escherichia coli , Anticuerpos/química , Anticuerpos/inmunología
5.
Anal Chem ; 96(24): 10003-10012, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38853531

RESUMEN

Fc-fusion proteins are an emerging class of protein therapeutics that combine the properties of biological ligands with the unique properties of the fragment crystallizable (Fc) domain of an immunoglobulin G (IgG). Due to their diverse higher-order structures (HOSs), Fc-fusion proteins remain challenging characterization targets within biopharmaceutical pipelines. While high-resolution biophysical tools are available for HOS characterization, they frequently demand extended time frames and substantial quantities of purified samples, rendering them impractical for swiftly screening candidate molecules. Herein, we describe the development of ion mobility-mass spectrometry (IM-MS) and collision-induced unfolding (CIU) workflows that aim to fill this technology gap, where we focus on probing the HOS of a model Fc-Interleukin-10 (Fc-IL-10) fusion protein engineered using flexible glycine-serine linkers. We evaluate the ability of these techniques to probe the flexibility of Fc-IL-10 in the absence of bulk solvent relative to other proteins of similar size, as well as localize structural changes of low charge state Fc-IL-10 ions to specific Fc and IL-10 unfolding events during CIU. We subsequently apply these tools to probe the local effects of glycine-serine linkers on the HOS and stability of IL-10 homodimer, which is the biologically active form of IL-10. Our data reveals that Fc-IL-10 produces significantly more structural transitions during CIU and broader IM profiles when compared to a wide range of model proteins, indicative of its exceptional structural dynamism. Furthermore, we use a combination of enzymatic approaches to annotate these intricate CIU data and localize specific transitions to the unfolding of domains within Fc-IL-10. Finally, we detect a strong positive, quadratic relationship between average linker mass and fusion protein stability, suggesting a cooperative influence between glycine-serine linkers and overall fusion protein stability. This is the first reported study on the use of IM-MS and CIU to characterize HOS of Fc-fusion proteins, illustrating the practical applicability of this approach.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Espectrometría de Masas , Desplegamiento Proteico , Proteínas Recombinantes de Fusión , Fragmentos Fc de Inmunoglobulinas/química , Proteínas Recombinantes de Fusión/química , Espectrometría de Masas/métodos , Interleucina-10/química , Interleucina-10/metabolismo , Espectrometría de Movilidad Iónica/métodos , Estabilidad Proteica , Humanos , Inmunoglobulina G/química
6.
Anal Biochem ; 693: 115585, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38851475

RESUMEN

Over the past few years, the implementation of mass spectrometry (MS) in QC laboratories has become a more common occurrence. The multi-attribute method (MAM), and emerging intact multi-attribute method (iMAM), are powerful analytical tools utilising liquid chromatography-mass spectrometry (LC-MS) methods that enable the monitoring of critical quality attributes (CQAs) in biotherapeutic proteins in compliant settings. Both MAM and iMAM are intended to replace or supplement several conventional assays with a single LC-MS method utilising MS data in combination with robust, semi-automated data processing workflows. MAM and iMAM workflows can also be implemented into current Good Manufacturing Practices environments due to the availability of CFR 11 compliant chromatography data system software. In this study, MAM and iMAM are employed for the analysis of 4 batches of a glucagon-like peptide-Fc fusion protein. MAM approach involved a first the discovery phase for the identification of CQAs and second, the target monitoring phase of the selected CQAs in other samples. New peak detection was performed on the data set to determine the appearance, absence or change of any peak. For native iMAM workflow both size exclusion and strong cation exchange chromatography were optimized for the identification and monitoring of CQAs at the intact level.


Asunto(s)
Proteínas Recombinantes de Fusión , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Flujo de Trabajo , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/análisis , Glucagón/análisis , Glucagón/química , Humanos , Espectrometría de Masas en Tándem/métodos , Cromatografía Líquida con Espectrometría de Masas
7.
Anal Methods ; 16(24): 3917-3926, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38832468

RESUMEN

The titer of recombinant proteins is one of the key parameters in biopharmaceutical manufacturing processes. The fluorescence polarization (FP)-based assay, a homogeneous, high-throughput and real-time analytical method, had emerged as a powerful tool for biochemical analysis and environmental monitoring. In this study, an FP-based bioassay was utilized to quantify antibody fragment crystallizable (Fc)-containing proteins, such as recombinant monoclonal antibodies (mAbs) and mAb derivatives, in the cell culture supernatant, and the impacts of tracer molecular weight and FITC-coupling conditions on fluorescence polarization were methodically examined. Distinct from the fluorescence polarization potency calculated by classical formula, we for the first time proposed a new concept and calculation of fluorescence polarization intensity, based on which an analytical method with broader detection range and analysis window was established for quantifying Fc-containing proteins. This provided new ideas for the practical application of fluorescence polarization theory. The established method could detect 96 samples within 30 minutes, with dynamic titer range of 2.5-400 mg L-1, and a linear fitting R2 between the measured and actual concentration reaching 0.99. The method had great application prospects in determining the titer of recombinant proteins with Fc fragments, especially when applied to large-scale screening of high-yield and stable expression CHO cell lines commonly used in biopharmaceutical industry.


Asunto(s)
Anticuerpos Monoclonales , Cricetulus , Polarización de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Fragmentos Fc de Inmunoglobulinas , Proteínas Recombinantes , Proteínas Recombinantes/química , Proteínas Recombinantes/análisis , Células CHO , Polarización de Fluorescencia/métodos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Ensayos Analíticos de Alto Rendimiento/métodos , Fragmentos Fc de Inmunoglobulinas/química , Bioensayo/métodos , Animales
8.
Sci Rep ; 14(1): 14832, 2024 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937649

RESUMEN

The structures of the Fc base of various IgG antibodies have been examined with a view to understanding how this region can be used to conjugate IgG to nanoparticles. The base structure is found to be largely consistent across a range of species and subtypes, comprising a hydrophobic region surrounded by hydrophilic residues, some of which are charged at physiological conditions. In addition, atomistic Molecular Dynamics simulations were performed to explore how model nanoparticles interact with the base using neutral and negatively charged gold nanoparticles. Both types of nanoparticle interacted readily with the base, leading to an adaptation of the antibody base surface to enhance the interactions. Furthermore, these interactions left the rest of the domain at the base of the Fc region structurally intact. This implies that coupling nanoparticles to the base of an IgG molecule is both feasible and desirable, since it leaves the antibody free to interact with its surroundings so that antigen-binding functionality can be retained. These results will therefore help guide future attempts to develop new nanotechnologies that exploit the unique properties of both antibodies and nanoparticles.


Asunto(s)
Oro , Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Nanopartículas del Metal , Simulación de Dinámica Molecular , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Fragmentos Fc de Inmunoglobulinas/química , Oro/química , Nanopartículas del Metal/química , Humanos , Nanopartículas/química , Interacciones Hidrofóbicas e Hidrofílicas , Animales
9.
Protein Expr Purif ; 220: 106503, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38759705

RESUMEN

Protein A affinity chromatography has been widely used for initial product capture in recombinant antibody/Fc-fusion purification. However, in general Protein A lacks the capability of separating aggregates (unless the aggregates are too large to enter the pores of resin beads or have their Protein A binding sites buried, in which case the aggregates do not bind). In the current work, we demonstrated that CaptureSelect FcXP affinity medium exhibited strong aggregate separation capability and effectively removed aggregates under pH or conductivity gradient elution in two bispecific antibody (bsAb) cases. For these two cases, aggregate contents were reduced from >16% and >22% (in the feed) to <1% and <5% (in the eluate) for the first and second bsAbs, respectively. While more case studies are required to further demonstrate FcXP's superiority in aggregate removal, findings from the current study suggest that FcXP can potentially be a better alternative than Protein A for product capture in cases where aggregate content is high.


Asunto(s)
Anticuerpos Biespecíficos , Cromatografía de Afinidad , Proteína Estafilocócica A , Cromatografía de Afinidad/métodos , Anticuerpos Biespecíficos/química , Anticuerpos Biespecíficos/aislamiento & purificación , Proteína Estafilocócica A/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/genética , Agregado de Proteínas , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/aislamiento & purificación
10.
Antiviral Res ; 227: 105917, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38782067

RESUMEN

The Fc-fused receptor binding domain (RBD-Fc) vaccine for SARS-CoV-2 has garnered significant attention for its capacity to provide effective and specific immune protection. However, its immunogenicity is limited, highlighting the need for improvement in clinical application. Nanoparticle delivery has been shown to be an effective method for enhancing antigen immunogenicity. In this study, we developed bivalent nanoparticle recombinant protein vaccines by assembling the RBD-Fc of SARS-CoV-2 and Fc-binding homo-oligomers o42.1 and i52.3 into octahedral and icosahedral nanoparticles. The formation of RBD-Fc nanoparticles was confirmed through structural characterization and cell binding experiments. Compared to RBD-Fc dimers, the nanoparticle vaccines induced more potent neutralizing antibodies (nAb) and stronger cellular immune responses. Therefore, using bivalent nanoparticle vaccines based on RBD-Fc presents a promising vaccination strategy against SARS-CoV-2 and offers a universal approach for enhancing the immunogenicity of Fc fusion protein vaccines.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Fragmentos Fc de Inmunoglobulinas , Nanovacunas , Glicoproteína de la Espiga del Coronavirus , Animales , Femenino , Humanos , Ratones , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/prevención & control , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Inmunidad Celular , Inmunogenicidad Vacunal , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/química , Ratones Endogámicos BALB C , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Desarrollo de Vacunas , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/química
11.
Antiviral Res ; 227: 105901, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38734211

RESUMEN

Growing concerns regarding the emergence of highly transmissible viral diseases highlight the urgent need to expand the repertoire of antiviral therapeutics. For this reason, new strategies for neutralizing and inhibiting these viruses are necessary. A promising approach involves targeting the glycans present on the surfaces of enveloped viruses. Lectins, known for their ability to recognize specific carbohydrate molecules, offer the potential for glycan-targeted antiviral strategies. Indeed, numerous studies have reported the antiviral effects of various lectins of both endogenous and exogenous origins. However, many lectins in their natural forms, are not suitable for use as antiviral therapeutics due to toxicity, other unfavorable pharmacological effects, and/or unreliable manufacturing sources. Therefore, improvements are crucial for employing lectins as effective antiviral therapeutics. A novel approach to enhance lectins' suitability as pharmaceuticals could be the generation of recombinant lectin-Fc fusion proteins, termed "lectibodies." In this review, we discuss the scientific rationale behind lectin-based antiviral strategies and explore how lectibodies could facilitate the development of new antiviral therapeutics. We will also share our perspective on the potential of these molecules to transcend their potential use as antiviral agents.


Asunto(s)
Antivirales , Lectinas , Antivirales/farmacología , Humanos , Lectinas/farmacología , Animales , Virosis/tratamiento farmacológico , Polisacáridos/química , Polisacáridos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/química , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/química , Virus/efectos de los fármacos
12.
J Chromatogr A ; 1726: 464947, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38724406

RESUMEN

Monoclonal antibodies (mAbs) are large and highly heterogeneous species typically characterized using a plethora of analytical methodologies. There is a trend within the biopharmaceutical industry to combine several of these methods in one analytical platform to simultaneously assess multiple structural attributes. Here, a protein analyzer for the fully automated middle-up and bottom-up liquid chromatography-mass spectrometry (LC-MS) analysis of charge, size and hydrophobic variants is described. The multidimensional set-up combines a multi-method option in the first dimension (1D) (choice between size exclusion - SEC, cation exchange - CEX or hydrophobic interaction chromatography - HIC) with second dimension (2D) on-column reversed-phase (RPLC) based desalting, denaturation and reduction prior to middle-up LC-MS analysis of collected 1D peaks and parallel on-column trypsin digestion of denatured and reduced peaks in the third dimension (3D) followed by bottom-up LC-MS analysis in the fourth dimension (4D). The versatile and comprehensive workflow is applied to the characterization of charge, hydrophobic and size heterogeneities associated with an engineered Fc fragment and is complemented with hydrogen-deuterium exchange (HDX) MS and FcRn affinity chromatography - native MS to explain observations in a structural/functional context.


Asunto(s)
Anticuerpos Monoclonales , Interacciones Hidrofóbicas e Hidrofílicas , Espectrometría de Masas , Anticuerpos Monoclonales/química , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Fragmentos Fc de Inmunoglobulinas/química , Humanos , Cromatografía en Gel/métodos , Cromatografía Líquida con Espectrometría de Masas
13.
PLoS One ; 19(4): e0300964, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38557973

RESUMEN

Human immunoglobulin G (IgG) exists as four subclasses IgG1-4, each of which has two Fab subunits joined by two hinges to a Fc subunit. IgG4 has the shortest hinge with 12 residues. The Fc subunit has two glycan chains, but the importance of glycosylation is not fully understood in IgG4. Here, to evaluate the stability and structure of non-glycosylated IgG4, we performed a multidisciplinary structural study of glycosylated and deglycosylated human IgG4 A33 for comparison with our similar study of human IgG1 A33. After deglycosylation, IgG4 was found to be monomeric by analytical ultracentrifugation; its sedimentation coefficient of 6.52 S was reduced by 0.27 S in reflection of its lower mass. X-ray and neutron solution scattering showed that the overall Guinier radius of gyration RG and its cross-sectional values after deglycosylation were almost unchanged. In the P(r) distance distribution curves, the two M1 and M2 peaks that monitor the two most common distances within IgG4 were unchanged following deglycosylation. Further insight from Monte Carlo simulations for glycosylated and deglycosylated IgG4 came from 111,382 and 117,135 possible structures respectively. Their comparison to the X-ray and neutron scattering curves identified several hundred best-fit models for both forms of IgG4. Principal component analyses showed that glycosylated and deglycosylated IgG4 exhibited different conformations from each other. Within the constraint of unchanged RG and M1-M2 values, the glycosylated IgG4 models showed more restricted Fc conformations compared to deglycosylated IgG4, but no other changes. Kratky plots supported this interpretation of greater disorder upon deglycosylation, also observed in IgG1. Overall, these more variable Fc conformations may demonstrate a generalisable impact of deglycosylation on Fc structures, but with no large conformational changes in IgG4 unlike those seen in IgG1.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Humanos , Inmunoglobulina G/química , Estudios Transversales , Modelos Moleculares , Fragmentos Fc de Inmunoglobulinas/química
14.
Nat Commun ; 15(1): 3600, 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38678029

RESUMEN

Streptococcus pyogenes can cause invasive disease with high mortality despite adequate antibiotic treatments. To address this unmet need, we have previously generated an opsonic IgG1 monoclonal antibody, Ab25, targeting the bacterial M protein. Here, we engineer the IgG2-4 subclasses of Ab25. Despite having reduced binding, the IgG3 version promotes stronger phagocytosis of bacteria. Using atomic simulations, we show that IgG3's Fc tail has extensive movement in 3D space due to its extended hinge region, possibly facilitating interactions with immune cells. We replaced the hinge of IgG1 with four different IgG3-hinge segment subclasses, IgGhxx. Hinge-engineering does not diminish binding as with IgG3 but enhances opsonic function, where a 47 amino acid hinge is comparable to IgG3 in function. IgGh47 shows improved protection against S. pyogenes in a systemic infection mouse model, suggesting that IgGh47 has promise as a preclinical therapeutic candidate. Importantly, the enhanced opsonic function of IgGh47 is generalizable to diverse S. pyogenes strains from clinical isolates. We generated IgGh47 versions of anti-SARS-CoV-2 mAbs to broaden the biological applicability, and these also exhibit strongly enhanced opsonic function compared to the IgG1 subclass. The improved function of the IgGh47 subclass in two distant biological systems provides new insights into antibody function.


Asunto(s)
COVID-19 , Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , SARS-CoV-2 , Streptococcus pyogenes , Animales , Humanos , Ratones , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , COVID-19/virología , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Ratones Endogámicos BALB C , Fagocitosis , Ingeniería de Proteínas/métodos , SARS-CoV-2/inmunología , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/inmunología
15.
J Biol Chem ; 300(5): 107245, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38569940

RESUMEN

The IgG-specific endoglycosidases EndoS and EndoS2 from Streptococcus pyogenes can remove conserved N-linked glycans present on the Fc region of host antibodies to inhibit Fc-mediated effector functions. These enzymes are therefore being investigated as therapeutics for suppressing unwanted immune activation, and have additional application as tools for antibody glycan remodeling. EndoS and EndoS2 differ in Fc glycan substrate specificity due to structural differences within their catalytic glycosyl hydrolase domains. However, a chimeric EndoS enzyme with a substituted glycosyl hydrolase from EndoS2 loses catalytic activity, despite high structural homology between the two enzymes, indicating either mechanistic divergence of EndoS and EndoS2, or improperly-formed domain interfaces in the chimeric enzyme. Here, we present the crystal structure of the EndoS2-IgG1 Fc complex determined to 3.0 Å resolution. Comparison of complexed and unliganded EndoS2 reveals relative reorientation of the glycosyl hydrolase, leucine-rich repeat and hybrid immunoglobulin domains. The conformation of the complexed EndoS2 enzyme is also different when compared to the earlier EndoS-IgG1 Fc complex, and results in distinct contact surfaces between the two enzymes and their Fc substrate. These findings indicate mechanistic divergence of EndoS2 and EndoS. It will be important to consider these differences in the design of IgG-specific enzymes, developed to enable customizable antibody glycosylation.


Asunto(s)
Proteínas Bacterianas , Glicósido Hidrolasas , Inmunoglobulina G , Modelos Moleculares , Streptococcus pyogenes , Humanos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Glicósido Hidrolasas/química , Glicósido Hidrolasas/metabolismo , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Streptococcus pyogenes/enzimología , Especificidad por Sustrato , Estructura Cuaternaria de Proteína
16.
J Pharm Biomed Anal ; 244: 116120, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38547650

RESUMEN

Charge heterogeneity is inherent to all therapeutic antibodies and arises from post-translational modifications (PTMs) and/or protein degradation events that may occur during manufacturing. Among therapeutic antibodies, the bispecific antibody (bsAb) containing two unique Fab arms directed against two different targets presents an additional layer of complexity to the charge profile. In the context of a bsAb, a single domain-specific PTM within one of the Fab domains may be sufficient to compromise target binding and could potentially impact the stability, safety, potency, and efficacy of the drug product. Therefore, characterization and routine monitoring of domain-specific modifications is critical to ensure the quality of therapeutic bispecific antibody products. We developed a Digestion-assisted imaged Capillary isoElectric focusing (DiCE) method to detect and quantitate domain-specific charge variants of therapeutic bispecific antibodies (bsAbs). The method involves enzymatic digestion using immunoglobulin G (IgG)-degrading enzyme of S. pyogenes (IdeS) to generate F(ab)2 and Fc fragments, followed by imaged capillary isoelectric focusing (icIEF) under reduced, denaturing conditions to separate the light chains (LCs) from the Fd domains. Our results suggest that DiCE is a highly sensitive method that is capable of quantitating domain-specific PTMs of a bsAb. In one case study, DiCE was used to quantitate unprocessed C-terminal lysine and site-specific glycation of Lys98 in the complementarity-determining region (CDR) of a bsAb that could not be accurately quantitated using conventional, platform-based charge variant analysis, such as intact icIEF. Quantitation of these PTMs by DiCE was comparable to results from peptide mapping, demonstrating that DiCE is a valuable orthogonal method for ensuring product quality. This method may also have potential applications for characterizing fusion proteins, antibody-drug conjugates, and co-formulated antibody cocktails.


Asunto(s)
Anticuerpos Biespecíficos , Focalización Isoeléctrica , Procesamiento Proteico-Postraduccional , Anticuerpos Biespecíficos/inmunología , Focalización Isoeléctrica/métodos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Humanos , Inmunoglobulina G/inmunología , Fragmentos Fc de Inmunoglobulinas/química
17.
Nat Protoc ; 19(6): 1887-1909, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38383719

RESUMEN

Immunoglobulin G (IgG) fragment crystallizable (Fc) glycosylation modulates effector functions such as antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. Consequently, assessing IgG Fc glycosylation is important for understanding the role of antibodies in infectious, alloimmune and autoimmune diseases. GlYcoLISA determines the Fc glycosylation of antigen-specific IgG by an immunosorbent assay with a liquid chromatography-mass spectrometry (LC-MS) readout. Detection of antigen-specific IgG glycosylation in a subclass- and site-specific manner is realized by LC-MS-based glycopeptide analysis after proteolytic cleavage. GlYcoLISA addresses challenges related to the low abundance of specific IgG and the high background of total IgG by using well-established immunosorbent assays for purifying antibodies of the desired specificity using immobilized antigen. Alternative methods with sufficient glycan resolution lack these important specificities. GlYcoLISA is performed in a 96-well plate format, and the analysis of 160 samples takes ~5 d, with 1 d for sample preparation, 2 d of LC-MS measurement and 2 d for partially automated data processing. GlYcoLISA requires expertise in LC-MS operation and data processing.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Inmunoglobulina G , Humanos , Antígenos/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Glicoproteínas , Glicosilación , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Inmunoglobulina G/inmunología , Cromatografía Líquida con Espectrometría de Masas
18.
J Chem Inf Model ; 64(3): 785-798, 2024 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-38262973

RESUMEN

The allosteric modulation of the homodimeric H10-03-6 protein to glycan ligands L1 and L2, and the STAB19 protein to glycan ligands L3 and L4, respectively, has been studied by molecular dynamics simulations and free energy calculations. The results revealed that the STAB19 protein has a significantly higher affinity for L3 (-11.38 ± 2.32 kcal/mol) than that for L4 (-5.51 ± 1.92 kcal/mol). However, the combination of the H10-03-6 protein with glycan L2 (1.23 ± 6.19 kcal/mol) is energetically unfavorable compared with that of L1 (-13.96 ± 0.35 kcal/mol). Further, the binding of glycan ligands L3 and L4 to STAB19 would result in the significant closure of the two CH2 domains of the STAB19 conformation with the decrease of the centroid distances between the two CH2 domains compared with the H10-03-6/L1/L2 complex. The CH2 domain closure of STAB19 relates directly to the formation of new hydrogen bonds and hydrophobic interactions between the residues Ser239, Val240, Asp265, Glu293, Asn297, Thr299, Ser337, Asp376, Thr393, Pro395, and Pro396 in STAB19 and glycan ligands L3 and L4, which suggests that these key residues would contribute to the specific regulation of STAB19 to L3 and L4. In addition, the distance analysis revealed that the EF loop in the H10-03-6/L1/L2 model presents a high flexibility and partial disorder compared with the stabilized STAB19/L3/L4 complex. These results will be helpful in understanding the specific regulation through the asymmetric structural characteristics in the CH2 and CH3 domains of the H10-03-6 and STAB19 proteins.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas , Simulación de Dinámica Molecular , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Isotipos de Inmunoglobulinas , Conformación Molecular , Polisacáridos
19.
J Biomol NMR ; 78(1): 9-18, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37989910

RESUMEN

Despite the prevalence and importance of glycoproteins in human biology, methods for isotope labeling suffer significant limitations. Common prokaryotic platforms do not produce mammalian post-translation modifications that are essential to the function of many human glycoproteins, including immunoglobulin G1 (IgG1). Mammalian expression systems require complex media and thus introduce significant costs to achieve uniform labeling. Expression with Pichia is available, though expertise and equipment requirements surpass E. coli culture. We developed a system utilizing Saccharomyces cerevisiae, [13C]-glucose, and [15N]-ammonium chloride with complexity comparable to E. coli. Here we report two vectors for expressing the crystallizable fragment (Fc) of IgG1 for secretion into the culture medium, utilizing the ADH2 or DDI2 promoters. We also report a strategy to optimize the expression yield using orthogonal Taguchi arrays. Lastly, we developed two different media formulations, a standard medium which provides 86-92% 15N and 30% 13C incorporation into the polypeptide, or a rich medium which provides 98% 15N and 95% 13C incorporation as determined by mass spectrometry. This advance represents an expression and optimization strategy accessible to experimenters with the capability to grow and produce proteins for NMR-based experiments using E. coli.


Asunto(s)
Escherichia coli , Saccharomyces cerevisiae , Animales , Humanos , Resonancia Magnética Nuclear Biomolecular/métodos , Glicoproteínas/química , Fragmentos Fc de Inmunoglobulinas/química , Inmunoglobulina G/química , Mamíferos
20.
J Phys Chem B ; 127(39): 8344-8357, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37751332

RESUMEN

Monoclonal antibodies (mAbs) make up a major class of biotherapeutics with a wide range of clinical applications. Their physical stability can be affected by various environmental factors. For instance, an acidic pH can be encountered during different stages of the mAb manufacturing process, including purification and storage. Therefore, understanding the behavior of flexible mAb molecules in acidic solution environments will benefit the development of stable mAb products. This study used small-angle X-ray scattering (SAXS) and complementary biophysical characterization techniques to investigate the conformational flexibility and protein-protein interactions (PPI) of a model mAb molecule under near-neutral and acidic conditions. The study also characterized the interactions between Fab and Fc fragments under the same buffer conditions to identify domain-domain interactions. The results suggest that solution pH significantly influences mAb flexibility and thus could help mAbs remain physically stable by maximizing local electrostatic repulsions when mAbs become crowded in solution. Under acidic buffer conditions, both Fab and Fc contribute to the repulsive PPI observed among the full mAb at a low ionic strength. However, as ionic strength increases, hydrophobic interactions lead to the self-association of Fc fragments and, subsequently, could affect the aggregation state of the mAb.


Asunto(s)
Anticuerpos Monoclonales , Inmunoglobulina G , Anticuerpos Monoclonales/química , Dispersión del Ángulo Pequeño , Inmunoglobulina G/química , Difracción de Rayos X , Cloruro de Sodio , Ácidos , Fragmentos Fc de Inmunoglobulinas/química , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA