Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(41): e2300258120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37801475

RESUMEN

Despite much effort, antibody therapies for Alzheimer's disease (AD) have shown limited efficacy. Challenges to the rational design of effective antibodies include the difficulty of achieving specific affinity to critical targets, poor expression, and antibody aggregation caused by buried charges and unstructured loops. To overcome these challenges, we grafted previously determined sequences of fibril-capping amyloid inhibitors onto a camel heavy chain antibody scaffold. These sequences were designed to cap fibrils of tau, known to form the neurofibrillary tangles of AD, thereby preventing fibril elongation. The nanobodies grafted with capping inhibitors blocked tau aggregation in biosensor cells seeded with postmortem brain extracts from AD and progressive supranuclear palsy (PSP) patients. The tau capping nanobody inhibitors also blocked seeding by recombinant tau oligomers. Another challenge to the design of effective antibodies is their poor blood-brain barrier (BBB) penetration. In this study, we also designed a bispecific nanobody composed of a nanobody that targets a receptor on the BBB and a tau capping nanobody inhibitor, conjoined by a flexible linker. We provide evidence that the bispecific nanobody improved BBB penetration over the tau capping inhibitor alone after intravenous administration in mice. Our results suggest that the design of synthetic antibodies that target sequences that drive protein aggregation may be a promising approach to inhibit the prion-like seeding of tau and other proteins involved in AD and related proteinopathies.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos de Dominio Único , Parálisis Supranuclear Progresiva , Humanos , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Proteínas tau/metabolismo , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/metabolismo , Ovillos Neurofibrilares/metabolismo , Parálisis Supranuclear Progresiva/metabolismo , Anticuerpos/metabolismo , Encéfalo/metabolismo
2.
J Biol Chem ; 298(9): 102357, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35952760

RESUMEN

Strains of Clostridium perfringens produce a two-domain enterotoxin (CpE) that afflicts humans and domesticated animals, causing prevalent gastrointestinal illnesses. CpE's C-terminal domain (cCpE) binds cell surface receptors, followed by a restructuring of its N-terminal domain to form a membrane-penetrating ß-barrel pore, which is toxic to epithelial cells of the gut. The claudin family of membrane proteins are known receptors for CpE and also control the architecture and function of cell-cell contacts (tight junctions) that create barriers to intercellular molecular transport. CpE binding and assembly disables claudin barrier function and induces cytotoxicity via ß-pore formation, disrupting gut homeostasis; however, a structural basis of this process and strategies to inhibit the claudin-CpE interactions that trigger it are both lacking. Here, we used a synthetic antigen-binding fragment (sFab) library to discover two sFabs that bind claudin-4 and cCpE complexes. We established these sFabs' mode of molecular recognition and binding properties and determined structures of each sFab bound to claudin-4-cCpE complexes using cryo-EM. The structures reveal that the sFabs bind a shared epitope, but conform distinctly, which explains their unique binding equilibria. Mutagenesis of antigen/sFab interfaces observed therein result in binding changes, validating the structures, and uncovering the sFab's targeting mechanism. From these insights, we generated a model for CpE's claudin-bound ß-pore that predicted sFabs would not prevent cytotoxicity, which we then verified in vivo. Taken together, this work demonstrates the development and mechanism of claudin/cCpE-binding sFabs that provide a framework and strategy for obstructing claudin/CpE assembly to treat CpE-linked gastrointestinal diseases.


Asunto(s)
Claudinas , Enterotoxinas , Animales , Claudina-3/genética , Claudina-3/metabolismo , Claudina-4/genética , Claudina-4/metabolismo , Claudinas/metabolismo , Clostridium perfringens , Enterotoxinas/metabolismo , Epítopos/metabolismo , Humanos , Unión Proteica
3.
Biochemistry (Mosc) ; 87(Suppl 1): S146-S110, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35501993

RESUMEN

Functional Protein Engineering became the hallmark in biomolecule manipulation in the new millennium, building on and surpassing the underlying structural DNA manipulation and recombination techniques developed and employed in the last decades of 20th century. Because of their prominence in almost all biological processes, proteins represent extremely important targets for engineering enhanced or altered properties that can lead to improvements exploitable in healthcare, medicine, research, biotechnology, and industry. Synthetic protein structures and functions can now be designed on a computer and/or evolved using molecular display or directed evolution methods in the laboratory. This review will focus on the recent trends in protein engineering and the impact of this technology on recent progress in science, cancer- and immunotherapies, with the emphasis on the current achievements in basic protein research using synthetic antibody (sABs) produced by phage display pipeline in the Kossiakoff laboratory at the University of Chicago (KossLab). Finally, engineering of the highly specific binding modules, such as variants of Streptococcal protein G with ultra-high orthogonal affinity for natural and engineered antibody scaffolds, and their possible applications as a plug-and-play platform for research and immunotherapy will be described.


Asunto(s)
Bacteriófagos , Investigación Biomédica , Anticuerpos , Bacteriófagos/genética , Biotecnología/métodos , Biblioteca de Péptidos , Ingeniería de Proteínas/métodos , Proteínas
4.
J Biol Chem ; 295(30): 10153-10167, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32439801

RESUMEN

Agonist stimulation of G-protein-coupled receptors (GPCRs) typically leads to phosphorylation of GPCRs and binding to multifunctional proteins called ß-arrestins (ßarrs). The GPCR-ßarr interaction critically contributes to GPCR desensitization, endocytosis, and downstream signaling, and GPCR-ßarr complex formation can be used as a generic readout of GPCR and ßarr activation. Although several methods are currently available to monitor GPCR-ßarr interactions, additional sensors to visualize them may expand the toolbox and complement existing methods. We have previously described antibody fragments (FABs) that recognize activated ßarr1 upon its interaction with the vasopressin V2 receptor C-terminal phosphopeptide (V2Rpp). Here, we demonstrate that these FABs efficiently report the formation of a GPCR-ßarr1 complex for a broad set of chimeric GPCRs harboring the V2R C terminus. We adapted these FABs to an intrabody format by converting them to single-chain variable fragments and used them to monitor the localization and trafficking of ßarr1 in live cells. We observed that upon agonist simulation of cells expressing chimeric GPCRs, these intrabodies first translocate to the cell surface, followed by trafficking into intracellular vesicles. The translocation pattern of intrabodies mirrored that of ßarr1, and the intrabodies co-localized with ßarr1 at the cell surface and in intracellular vesicles. Interestingly, we discovered that intrabody sensors can also report ßarr1 recruitment and trafficking for several unmodified GPCRs. Our characterization of intrabody sensors for ßarr1 recruitment and trafficking expands currently available approaches to visualize GPCR-ßarr1 binding, which may help decipher additional aspects of GPCR signaling and regulation.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 1/metabolismo , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Transporte de Proteínas , Receptores Acoplados a Proteínas G/genética , beta-Arrestina 1/genética
5.
Chembiochem ; 22(9): 1589-1596, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32964656

RESUMEN

De novo cancer-targeting immunostimulatory peptides have been designed and developed as synthetic antibody mimics. A series of bifunctional peptides incorporating NKp30-binding and NK-cell-activating domains were synthesized as linear dimers and then extended into branching trimeric peptides by the incorporation of GRP78-targeting and tumor-cell-binding sequences. A selected trimeric peptide from this small set of peptides displayed binding capabilities on GRP78+ HepG2 and A549 target cells. Cell binding diminished in the presence of an anti-GRP78 peptide blocker, thus suggesting GRP78-binding dependence. Similarly, the selected trimeric peptide was also found to exhibit NK cell binding in an NKp30-dependent manner, which translated into NK cell activation as indicated by cytokine secretion. In co-culture, fluorescence microscopy revealed that the target GFP-expressing A549 cells were visibly associated with the effector NK cells when pre-activated with lead trimeric peptide. Accordingly, A549 cells were found to be compromised, as evidenced by the loss of GFP signal and notable detection of early-/late-stage apoptosis. Investigation of the immunological markers related to toxicity revealed detectable secretion of pro-inflammatory cytokines and chemokines, including IFN-γ, TNF-α, and IL-8. Furthermore, administration of peptide-activated NK cells into A549-tumor-bearing mice resulted in a consistent decrease in tumor growth when compared to the untreated control group. Taken together, the identification of a lead trimeric peptide capable of targeting and activating NK cells' immunotoxicity directly towards GRP78+ /B7H6- tumors provides a novel proof-of-concept for the development of cancer-targeting immunostimulatory peptide ligands that mimic antibody-targeting and -activating functions related to cancer immunotherapy applications.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Anticuerpos/química , Células Asesinas Naturales/efectos de los fármacos , Péptidos/química , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/uso terapéutico , Animales , Anticuerpos/inmunología , Línea Celular Tumoral , Citocinas/metabolismo , Chaperón BiP del Retículo Endoplásmico/inmunología , Femenino , Humanos , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Péptidos/síntesis química , Péptidos/farmacología , Péptidos/uso terapéutico , Trasplante Heterólogo
6.
Fish Shellfish Immunol ; 86: 361-367, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30502461

RESUMEN

The emerging technology of aptamers that is also known as synthetic antibodies is rivalling antibodies research in the recent years. The unique yet important features of aptamers are advancing antibodies in diverse applications, which include disease diagnosis, prophylactic and therapeutic. The versatility of aptamer has further extended its application to function as gene expression modulator, known as synthetic riboswitches. This report reviewed and discussed the applications of aptamers technology in the biosecurity of aquaculture, the promising developments in biosensor detection for disease diagnosis as well as prophylactic and therapeutic measurements. The application of aptamers technology in immunophenotyping study of aquatic animal is highlighted. Lastly, the future perspective of aptamers in the management of aquatic animal health is discussed, special emphasis on the potential application of aptamers as synthetic riboswitches to enhance host immunity, as well as the growth performance.


Asunto(s)
Anticuerpos/inmunología , Acuicultura , Inmunofenotipificación/veterinaria , Oligonucleótidos/inmunología , Animales , Inmunofenotipificación/métodos
7.
Biotechnol Lett ; 41(1): 79-90, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30357542

RESUMEN

OBJECTIVES: Designing a polypeptide sequence to interact with a preselected target polypeptide sequence of a protein has long been of interest, yet remains an elusive goal. RESULTS: Here, we propose a novel concept named "Clustered Complementary Amino Acid Pairing (CCAAP)," which plays an essential role in protein-protein interaction (PPI). Complementary amino acid pairing (CAAP) is a pairing between two amino acids encoded by a codon and its reverse complementary codon. CAAP interactions largely agree with the physicochemical and stereochemical requirements for probable amino acid pairings. Interestingly, 82 PPI structure data revealed that clusters of CAAP interactions (CCAAP boxes) are predominantly found in all PPI sites. Analysis of all amino acid pairings in the CCAAP boxes unveiled amino acid-pairing preferences and patterns for PPI that allowed us to develop a new method for designing an oligopeptide sequence to bind to a chosen polypeptide sequence of any target protein. CONCLUSIONS: Discoveries in the present study provide proof of the CCAAP principle.


Asunto(s)
Codón/genética , Oligopéptidos/genética , Análisis de Secuencia de Proteína , Oligopéptidos/química
8.
RNA ; 22(4): 636-55, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26847261

RESUMEN

Post-transcriptional regulation of mRNAs plays an essential role in the control of gene expression. mRNAs are regulated in ribonucleoprotein (RNP) complexes by RNA-binding proteins (RBPs) along with associated protein and noncoding RNA (ncRNA) cofactors. A global understanding of post-transcriptional control in any cell type requires identification of the components of all of its RNP complexes. We have previously shown that these complexes can be purified by immunoprecipitation using anti-RBP synthetic antibodies produced by phage display. To develop the large number of synthetic antibodies required for a global analysis of RNP complex composition, we have established a pipeline that combines (i) a computationally aided strategy for design of antigens located outside of annotated domains, (ii) high-throughput antigen expression and purification in Escherichia coli, and (iii) high-throughput antibody selection and screening. Using this pipeline, we have produced 279 antibodies against 61 different protein components of Drosophila melanogaster RNPs. Together with those produced in our low-throughput efforts, we have a panel of 311 antibodies for 67 RNP complex proteins. Tests of a subset of our antibodies demonstrated that 89% immunoprecipitate their endogenous target from embryo lysate. This panel of antibodies will serve as a resource for global studies of RNP complexes in Drosophila. Furthermore, our high-throughput pipeline permits efficient production of synthetic antibodies against any large set of proteins.


Asunto(s)
Anticuerpos/química , Proteínas de Drosophila/inmunología , Ribonucleoproteínas/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos/metabolismo , Antígenos/inmunología , Antígenos/aislamiento & purificación , Western Blotting , Regiones Determinantes de Complementariedad , Proteínas de Drosophila/aislamiento & purificación , Drosophila melanogaster , Ensayo de Inmunoadsorción Enzimática , Escherichia coli , Inmunoprecipitación , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Ribonucleoproteínas/aislamiento & purificación
9.
J Biol Chem ; 291(1): 89-102, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26565029

RESUMEN

The BCL-2 protein family plays a critical role in regulating cellular commitment to mitochondrial apoptosis. Pro-apoptotic Bcl-2-associated X protein (BAX) is an executioner protein of the BCL-2 family that represents the gateway to mitochondrial apoptosis. Following cellular stresses that induce apoptosis, cytosolic BAX is activated and translocates to the mitochondria, where it inserts into the mitochondrial outer membrane to form a toxic pore. How the BAX activation pathway proceeds and how this may be inhibited is not yet completely understood. Here we describe synthetic antibody fragments (Fabs) as structural and biochemical probes to investigate the potential mechanisms of BAX regulation. These synthetic Fabs bind with high affinity to BAX and inhibit its activation by the BH3-only protein tBID (truncated Bcl2 interacting protein) in assays using liposomal membranes. Inhibition of BAX by a representative Fab, 3G11, prevented mitochondrial translocation of BAX and BAX-mediated cytochrome c release. Using NMR and hydrogen-deuterium exchange mass spectrometry, we showed that 3G11 forms a stoichiometric and stable complex without inducing a significant conformational change on monomeric and inactive BAX. We identified that the Fab-binding site on BAX involves residues of helices α1/α6 and the α1-α2 loop. Therefore, the inhibitory binding surface of 3G11 overlaps with the N-terminal activation site of BAX, suggesting a novel mechanism of BAX inhibition through direct binding to the BAX N-terminal activation site. The synthetic Fabs reported here reveal, as probes, novel mechanistic insights into BAX inhibition and provide a blueprint for developing inhibitors of BAX activation.


Asunto(s)
Anticuerpos/farmacología , Proteína X Asociada a bcl-2/antagonistas & inhibidores , Proteína X Asociada a bcl-2/química , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Sitios de Unión , Citocromos c/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/metabolismo , Liposomas/metabolismo , Espectroscopía de Resonancia Magnética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Permeabilidad/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
10.
Biopolymers ; 108(2)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27539157

RESUMEN

We report on peptide-based ligands matured through the protein catalyzed capture (PCC) agent method to tailor molecular binders for in vitro sensing/diagnostics and in vivo pharmacokinetics parameters. A vascular endothelial growth factor (VEGF) binding peptide and a peptide against the protective antigen (PA) protein of Bacillus anthracis discovered through phage and bacterial display panning technologies, respectively, were modified with click handles and subjected to iterative in situ click chemistry screens using synthetic peptide libraries. Each azide-alkyne cycloaddition iteration, promoted by the respective target proteins, yielded improvements in metrics for the application of interest. The anti-VEGF PCC was explored as a stable in vivo imaging probe. It exhibited excellent stability against proteases and a mean elimination in vivo half-life (T1/2 ) of 36 min. Intraperitoneal injection of the reagent results in slow clearance from the peritoneal cavity and kidney retention at extended times, while intravenous injection translates to rapid renal clearance. The ligand competed with the commercial antibody for binding to VEGF in vivo. The anti-PA ligand was developed for detection assays that perform in demanding physical environments. The matured anti-PA PCC exhibited no solution aggregation, no fragmentation when heated to 100°C, and > 81% binding activity for PA after heating at 90°C for 1 h. We discuss the potential of the PCC agent screening process for the discovery and enrichment of next generation antibody alternatives.


Asunto(s)
Química Clic/métodos , Biblioteca de Péptidos , Péptidos/química , Factor A de Crecimiento Endotelial Vascular/química , Secuencia de Aminoácidos , Animales , Anticuerpos/administración & dosificación , Anticuerpos/química , Anticuerpos/metabolismo , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Rastreo Diferencial de Calorimetría , Catálisis , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/metabolismo , Femenino , Células HT29 , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Ligandos , Masculino , Espectrometría de Masas , Ratones , Microsomas Hepáticos/metabolismo , Péptidos/metabolismo , Péptidos/farmacocinética , Unión Proteica , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Bioorg Med Chem ; 25(18): 5008-5015, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28583807

RESUMEN

The synthesis of a semi-orthogonally protected CycloTriVeratrilene (CTV) scaffold derivative as well as the sequential introduction of three different peptide loops onto this molecular scaffold via Cu(I)-catalyzed azide alkyne cycloaddition towards a medium-sized protein mimic is described. This approach for the construction of medium-sized protein mimics is illustrated by the synthesis of a paratope mimic of the monoclonal antibody Infliximab (Remicade®) and provides access to a range of highly pre-organized molecular constructs bearing three different peptide segments. This approach may find wide applications for development of protein-protein interaction disruptors as well as synthetic vaccines.


Asunto(s)
Péptidos Cíclicos/síntesis química , Compuestos Policíclicos/química , Alquinos/química , Azidas/química , Catálisis , Cobre/química , Reacción de Cicloadición , Infliximab/química , Infliximab/metabolismo , Péptidos Cíclicos/química
12.
Adv Exp Med Biol ; 1053: 79-98, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29549636

RESUMEN

Rapid spread of microbial resistance and recent outbreaks of viral disease have led to renewed interest in antibody-based therapies for infectious diseases. Synthetic antibody libraries displayed on phage offer unique advantages over traditional immunization-based antibody generation, including full control over library design and selection conditions. The technology has matured beyond natural antibodies and is capable of providing novel insights into infectious disease and can generate novel antibodies that cannot be produced by the natural immune system. This chapter gives an overview of recombinant antibody library technology with an emphasis on our work using a highly successful synthetic single framework Fab library. We demonstrate its utility in targeting viruses and bacterial toxins in five case studies.


Asunto(s)
Antiinfecciosos , Anticuerpos Monoclonales/genética , Técnicas de Visualización de Superficie Celular , Enfermedades Transmisibles/tratamiento farmacológico , Fragmentos Fab de Inmunoglobulinas/genética , Biblioteca de Péptidos , Animales , Antiinfecciosos/inmunología , Antiinfecciosos/uso terapéutico , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Enfermedades Transmisibles/inmunología , Interacciones Huésped-Patógeno , Humanos , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/uso terapéutico
13.
Adv Exp Med Biol ; 1053: 21-34, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29549633

RESUMEN

Antibody phage display has become an indispensable tool for the discovery and optimization of target-specific monoclonal antibodies suitable for demanding applications including therapeutic reagents. The in vitro nature of the technology enables the rapid and efficient identification of specific binders, as well as greater control over selection parameters that facilitates the isolation of antibodies with unique, desirable functional characteristics. In this chapter, the technological background and the state of the art in the field of antibody phage display is discussed.


Asunto(s)
Anticuerpos Monoclonales/genética , Bacteriófago M13/genética , Técnicas de Visualización de Superficie Celular , Escherichia coli/virología , Fragmentos Fab de Inmunoglobulinas/genética , Biblioteca de Péptidos , Anticuerpos de Cadena Única/genética , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/uso terapéutico
14.
BMC Bioinformatics ; 17: 150, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27039071

RESUMEN

BACKGROUND: The identification of structured units in a protein sequence is an important first step for most biochemical studies. Importantly for this study, the identification of stable structured region is a crucial first step to generate novel synthetic antibodies. While many approaches to find domains or predict structured regions exist, important limitations remain, such as the optimization of domain boundaries and the lack of identification of non-domain structured units. Moreover, no integrated tool exists to find and optimize structural domains within protein sequences. RESULTS: Here, we describe a new tool, PAT ( http://www.kimlab.org/software/pat ) that can efficiently identify both domains (with optimized boundaries) and non-domain putative structured units. PAT automatically analyzes various structural properties, evaluates the folding stability, and reports possible structural domains in a given protein sequence. For reliability evaluation of PAT, we applied PAT to identify antibody target molecules based on the notion that soluble and well-defined protein secondary and tertiary structures are appropriate target molecules for synthetic antibodies. CONCLUSION: PAT is an efficient and sensitive tool to identify structured units. A performance analysis shows that PAT can characterize structurally well-defined regions in a given sequence and outperforms other efforts to define reliable boundaries of domains. Specially, PAT successfully identifies experimentally confirmed target molecules for antibody generation. PAT also offers the pre-calculated results of 20,210 human proteins to accelerate common queries. PAT can therefore help to investigate large-scale structured domains and improve the success rate for synthetic antibody generation.


Asunto(s)
Anticuerpos/inmunología , Proteínas/química , Interfaz Usuario-Computador , Secuencia de Aminoácidos , Anticuerpos/química , Área Bajo la Curva , Bases de Datos de Proteínas , Humanos , Internet , Biblioteca de Péptidos , Estructura Terciaria de Proteína , Proteínas/inmunología , Curva ROC
15.
J Biol Chem ; 289(50): 34851-61, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25352592

RESUMEN

The γ-secretase complex, composed of presenilin, nicastrin (NCT), anterior pharynx-defective 1 (APH-1), and presenilin enhancer 2 (PEN-2), is assembled in a highly regulated manner and catalyzes the intramembranous proteolysis of many type I membrane proteins, including Notch and amyloid precursor protein. The Notch family of receptors plays important roles in cell fate specification during development and in adult tissues, and aberrant hyperactive Notch signaling causes some forms of cancer. γ-Secretase-mediated processing of Notch at the cell surface results in the generation of the Notch intracellular domain, which associates with several transcriptional coactivators involved in nuclear signaling events. On the other hand, γ-secretase-mediated processing of amyloid precursor protein leads to the production of amyloid ß (Aß) peptides that play an important role in the pathogenesis of Alzheimer disease. We used a phage display approach to identify synthetic antibodies that specifically target NCT and expressed them in the single-chain variable fragment (scFv) format in mammalian cells. We show that expression of a NCT-specific scFv clone, G9, in HEK293 cells decreased the production of the Notch intracellular domain but not the production of amyloid ß peptides that occurs in endosomal and recycling compartments. Biochemical studies revealed that scFvG9 impairs the maturation of NCT by associating with immature forms of NCT and, consequently, prevents its association with the other components of the γ-secretase complex, leading to degradation of these molecules. The reduced cell surface levels of mature γ-secretase complexes, in turn, compromise the intramembranous processing of Notch.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Anticuerpos de Cadena Única/inmunología , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/inmunología , Especificidad de Anticuerpos , Células HEK293 , Humanos , Espacio Intracelular/metabolismo , Glicoproteínas de Membrana/química , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteolisis , Receptores Notch/metabolismo , Anticuerpos de Cadena Única/genética
16.
Int J Biol Macromol ; 256(Pt 1): 128191, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38000614

RESUMEN

Nanobodies are natural anti-SARS-CoV-2 drug candidates. Engineering multivalent nanobodies is an effective way to improve the functional binding affinity of natural nanobodies by simultaneously targeting multiple sites on viral proteins. However, multivalent nanobodies have usually been engineered by trial and error, and rational designs are still lacking. Here, we describe a structure-guided design of a self-assembled trivalent nanobody cluster targeting the SARS-CoV-2 spike protein. Using the nanobody Nb6 as a monovalent binder, we first selected a human-derived trimerization scaffold evaluated by molecular dynamics simulations, then selected an optimal linker according to the minimum distance between Nb6 and the trimerization scaffold, and finally successfully engineered a trivalent nanobody cluster called Tribody. Compared with the low-affinity monovalent counterpart (Nb6), Tribody showed much higher target binding affinity (KD < 1 pM) and thus had a 900-fold increase in antiviral neutralization against SARS-CoV-2 pseudovirus. We determined the cryo-EM structure of the Tribody-spike complex and confirmed that all three Nb6 binders of Tribody collectively bind to the three receptor-binding domains (RBDs) of the spike and lock them in a 3-RBD-down conformation, fully consistent with our structure-guided design. This study demonstrates that synthetic nanobody clusters with human-derived self-assembled scaffolds are potential protein drugs against SARS-CoV-2 coronaviruses.


Asunto(s)
COVID-19 , Anticuerpos de Dominio Único , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Neutralizantes , Unión Proteica
17.
Mol Biotechnol ; 2024 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-38308755

RESUMEN

Synthetic antibodies (Abs) represent a category of engineered proteins meticulously crafted to replicate the functions of their natural counterparts. Such Abs are generated in vitro, enabling advanced molecular alterations associated with antigen recognition, paratope site engineering, and biochemical refinements. In a parallel realm, deep sequencing has brought about a paradigm shift in molecular biology. It facilitates the prompt and cost-effective high-throughput sequencing of DNA and RNA molecules, enabling the comprehensive big data analysis of Ab transcriptomes, including specific regions of interest. Significantly, the integration of artificial intelligence (AI), based on machine- and deep- learning approaches, has fundamentally transformed our capacity to discern patterns hidden within deep sequencing big data, including distinctive Ab features and protein folding free energy landscapes. Ultimately, current AI advances can generate approximations of the most stable Ab structural configurations, enabling the prediction of de novo synthetic Abs. As a result, this manuscript comprehensively examines the latest and relevant literature concerning the intersection of deep sequencing big data and AI methodologies for the design and development of synthetic Abs. Together, these advancements have accelerated the exploration of antibody repertoires, contributing to the refinement of synthetic Ab engineering and optimizations, and facilitating advancements in the lead identification process.

18.
Methods Mol Biol ; 2552: 437-445, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36346607

RESUMEN

To ensure the functionalities of the antibodies in phage-displayed synthetic antibody libraries, we use computational method to evaluate the designs of the antibody libraries. The computational methodologies developed in our lab for designing antibody library provide rich information on the function of the designed antibody sequences-adequate antibody designs for a specific antigen type should have predicted paratopes for the antigen type. This computational assessment of the designed antibody sequences helps eliminate non-functional designs before proceeding to construct the library designs in the wet lab. As such, only reasonable antibody designs are constructed for antibody discoveries.


Asunto(s)
Anticuerpos , Biblioteca de Péptidos , Sitios de Unión de Anticuerpos , Antígenos
19.
ACS Nano ; 17(7): 7017-7034, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-36971310

RESUMEN

The rapid emergence and spread of vaccine/antibody-escaping variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed serious challenges to our efforts in combating corona virus disease 2019 (COVID-19) pandemic. A potent and broad-spectrum neutralizing reagent against these escaping mutants is extremely important for the development of strategies for the prevention and treatment of SARS-CoV-2 infection. We herein report an abiotic synthetic antibody inhibitor as a potential anti-SARS-CoV-2 therapeutic agent. The inhibitor, Aphe-NP14, was selected from a synthetic hydrogel polymer nanoparticle library created by incorporating monomers with functionalities complementary to key residues of the SARS-CoV-2 spike glycoprotein receptor binding domain (RBD) involved in human angiotensin-converting enzyme 2 (ACE2) binding. It has high capacity, fast adsorption kinetics, strong affinity, and broad specificity in biologically relevant conditions to both the wild type and the current variants of concern, including Beta, Delta, and Omicron spike RBD. The Aphe-NP14 uptake of spike RBD results in strong blockage of spike RBD-ACE2 interaction and thus potent neutralization efficacy against these escaping spike protein variant pseudotyped viruses. It also inhibits live SARS-CoV-2 virus recognition, entry, replication, and infection in vitro and in vivo. The Aphe-NP14 intranasal administration is found to be safe due to its low in vitro and in vivo toxicity. These results establish a potential application of abiotic synthetic antibody inhibitors in the prevention and treatment of the infection of emerging or possibly future SARS-CoV-2 variants.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Polímeros , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Unión Proteica , Anticuerpos Antivirales , Glicoproteína de la Espiga del Coronavirus
20.
Front Immunol ; 14: 1089395, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37180155

RESUMEN

Background: Monoclonal antibodies (mAbs) and their derivatives are the fastest expanding category of pharmaceuticals. Efficient screening and generation of appropriate therapeutic human antibodies are important and urgent issues in the field of medicine. The successful in vitro biopanning method for antibody screening largely depends on the highly diverse, reliable and humanized CDR library. To rapidly obtain potent human antibodies, we designed and constructed a highly diverse synthetic human single-chain variable fragment (scFv) antibody library greater than a giga in size by phage display. Herein, the novel TIM-3-neutralizing antibodies with immunomodulatory functions derived from this library serve as an example to demonstrate the library's potential for biomedical applications. Methods: The library was designed with high stability scaffolds and six complementarity determining regions (CDRs) tailored to mimic human composition. The engineered antibody sequences were optimized for codon usage and subjected to synthesis. The six CDRs with variable length CDR-H3s were individually subjected to ß-lactamase selection and then recombined for library construction. Five therapeutic target antigens were used for human antibody generation via phage library biopanning. TIM-3 antibody activity was verified by immunoactivity assays. Results: We have designed and constructed a highly diverse synthetic human scFv library named DSyn-1 (DCB Synthetic-1) containing 2.5 × 1010 phage clones. Three selected TIM-3-recognizing antibodies DCBT3-4, DCBT3-19, and DCBT3-22 showed significant inhibition activity by TIM-3 reporter assays at nanomolar ranges and binding affinities in sub-nanomolar ranges. Furthermore, clone DCBT3-22 was exceptionally superior with good physicochemical property and a purity of more than 98% without aggregation. Conclusion: The promising results illustrate not only the potential of the DSyn-1 library for biomedical research applications, but also the therapeutic potential of the three novel fully human TIM-3-neutralizing antibodies.


Asunto(s)
Bacteriófagos , Anticuerpos de Cadena Única , Humanos , Biblioteca de Péptidos , Receptor 2 Celular del Virus de la Hepatitis A , Regiones Determinantes de Complementariedad/química , Anticuerpos Monoclonales , Anticuerpos de Cadena Única/genética , Anticuerpos Neutralizantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA