Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Anal Chim Acta ; 1264: 341300, 2023 Jul 11.
Article in English | MEDLINE | ID: covidwho-2316794

ABSTRACT

The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread worldwide which triggered serious public health issues. The search for rapid and accurate diagnosis, effective prevention, and treatment is urgent. The nucleocapsid protein (NP) of SARS-CoV-2 is one of the main structural proteins expressed and most abundant in the virus, and is considered a diagnostic marker for the accurate and sensitive detection of SARS-CoV-2. Herein, we report the screening of specific peptides from the pIII phage library that bind to SARS-CoV-2 NP. The phage monoclone expressing cyclic peptide N1 (peptide sequence, ACGTKPTKFC, with C&C bridged by disulfide bonding) specifically recognizes SARS-CoV-2 NP. Molecular docking studies reveal that the identified peptide is bound to the "pocket" region on the SARS-CoV-2 NP N-terminal domain mainly by forming a hydrogen bonding network and through hydrophobic interaction. Peptide N1 with the C-terminal linker was synthesized as the capture probe for SARS-CoV-2 NP in ELISA. The peptide-based ELISA was capable of assaying SARS-CoV-2 NP at concentrations as low as 61 pg/mL (∼1.2 pM). Furthermore, the as-proposed method could detect the SARS-CoV-2 virus at limits as low as 50 TCID50 (median tissue culture infective dose)/mL. This study demonstrates that selected peptides are powerful biomolecular tools for SARS-CoV-2 detection, providing a new and inexpensive method of rapidly screening infections as well as rapidly diagnosing coronavirus disease 2019 patients.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Bioprospecting , Molecular Docking Simulation , COVID-19/diagnosis , Nucleocapsid Proteins , Enzyme-Linked Immunosorbent Assay/methods , Peptides , Antibodies, Viral
2.
Fish Shellfish Immunol ; 138: 108807, 2023 Jul.
Article in English | MEDLINE | ID: covidwho-2316095

ABSTRACT

The COVID-19 pandemic has significantly impacted human health for three years. To mitigate the spread of SARS-CoV-2, the development of neutralizing antibodies has been accelerated, including the exploration of alternative antibody formats such as single-domain antibodies. In this study, we identified variable new antigen receptors (VNARs) specific for the receptor binding domain (RBD) of SARS-CoV-2 by immunizing a banded houndshark (Triakis scyllium) with recombinant wild-type RBD. Notably, the CoV2NAR-1 clone showed high binding affinities in the nanomolar range to various RBDs and demonstrated neutralizing activity against SARS-CoV-2 pseudoviruses. These results highlight the potential of the banded houndshark as an animal model for the development of VNAR-based therapeutics or diagnostics against future pandemics.


Subject(s)
COVID-19 , Single-Domain Antibodies , Humans , Animals , SARS-CoV-2/metabolism , Antibodies, Viral , Pandemics , Antibodies, Neutralizing
3.
J Biol Chem ; 299(6): 104831, 2023 06.
Article in English | MEDLINE | ID: covidwho-2315850

ABSTRACT

Viral proteases play key roles in viral replication, and they also facilitate immune escape by proteolyzing diverse target proteins. Deep profiling of viral protease substrates in host cells is beneficial for understanding viral pathogenesis and for antiviral drug discovery. Here, we utilized substrate phage display coupled with protein network analysis to identify human proteome substrates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteases, including papain-like protease (PLpro) and 3C-like protease (3CLpro). We first performed peptide substrates selection of PLpro and 3CLpro, and we then used the top 24 preferred substrate sequences to identify a total of 290 putative protein substrates. Protein network analysis revealed that the top clusters of PLpro and 3CLpro substrate proteins contain ubiquitin-related proteins and cadherin-related proteins, respectively. We verified that cadherin-6 and cadherin-12 are novel substrates of 3CLpro, and CD177 is a novel substrate of PLpro using in vitro cleavage assays. We thus demonstrated that substrate phage display coupled with protein network analysis is a simple and high throughput method to identify human proteome substrates of SARS-CoV-2 viral proteases for further understanding of virus-host interactions.


Subject(s)
COVID-19 , SARS-CoV-2 , Viral Proteases , Humans , Peptide Hydrolases/metabolism , Proteome , SARS-CoV-2/enzymology , SARS-CoV-2/metabolism
4.
Application of Natural Products in SARS-CoV-2 ; : 273-292, 2022.
Article in English | Scopus | ID: covidwho-2262170

ABSTRACT

The emerging human pathogenic viruses, including the recently emerged severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), have markedly affected the human health and have become a challenge for researchers worldwide. Antibiotic therapy and existing vaccines have reduced the pandemic burden to some extent. However, there is still need for efficient treatment, vaccination, and antiviral agents to control the pandemic. This chapter illustrates the role of bacteriophage in bacterial infections, SARS-COV-2 infected patients, biological activities of phages, phage display method, phages as potential inducers of antiviral immunity, phage-based vaccines, CRISPR and phage-based SARS-CoV-2 vaccines, and possible advantages of phage-based vaccines. It is concluded that phages have considerable breadth in the SARS-CoV-2 pandemic and offer many substantial advantages, such as clearing respiratory bacterial infections, which significantly reduce the burden of mortalities. Phage plays a vital role in triggering antiviral immunity by inducing cytokines such as IFN-α and IL-12. It suggests the role in driving antiviral immunity, triggering TLR3-dependent pattern recognition receptors, inhibiting TNF-driving type I IFN, inducing antiviral immunity through upregulation of the expression of defensin in IL-2, and encouraging a marked upregulation of gene hBD2 that induces virucidal effects, thus playing a key role in anti-SARS-COV-2 immunity. Moreover, phages have been presented as an alternative universal adjuvant-free nano-vaccine platform in which single-phage scaffolds are used to incorporate multiple targets. © 2023 Elsevier Inc. All rights reserved.

5.
European Respiratory Journal Conference: European Respiratory Society International Congress, ERS ; 60(Supplement 66), 2022.
Article in English | EMBASE | ID: covidwho-2259018

ABSTRACT

Background: In outpatients, monoclonal antibodies to Spike protein reduce viral load and improve outcomes, with a greater effect in serum antibody-negative at baseline. The aim of this study was to find epitope candidates to produce neutralizing monoclonal antibodies (mAb) for COVID-19 treatment. Method(s): IgG COVID-19 patients (N=500) against SARS-CoV-2 was confirmed. Epitope mapping was performed by Luminex technology. A computational pipeline based in predictive models was designed to predict S protein epitopes most likely to be recognized by mAb from COVID-19 convalescent patients. Result(s): Validation Screening: 29 epitopes of the SARS-CoV-2 S protein were predicted by our pipeline and included in the Luminex panel. 40 serum samples from convalescent COVID-19 patients and 126 pre-pandemia negative controls were included in the validation screening. Epitope mapping: 500 serum samples were tested against the 8 epitopes selected in the validation screening. The two epitopes with the highest IgG of participants above the seropositivity cut-offs were selected. The two most immunogenic epitopes were screened in phage library containing 109 clones of antibodies anti-SARS-CoV-2 to produce mAbs by phage display technology. Conclusion(s): The two epitopes with the highest IgG reactivity validated against serum samples from 500 COVID-19 convalescent patients and phage library are good candidates for the production of new neutralizing mAbs against SARS-CoV-2 S protein.

6.
Extreme Medicine ; - (4):5-12, 2022.
Article in English | Scopus | ID: covidwho-2250978

ABSTRACT

Monoclonal antibodies (mAbs) are the most promising and most intensively replenished type of bioactive pharmaceuticals. Currently, there are over 100 different mAbs approved by the FDA and other regulating agencies for treatment of oncological, infectious, systemic, autoimmune and other diseases. Design of antibodies neutralizing pathogens of socially significant infections, such as HIV, hepatitis viruses, SARS-CoV-2, is a separate direction. The SARS-CoV-2 pandemic has shown how urgent it is to have a technological platform enabling production of fully human antibodies. The development of recombinant DNA technology and antibody phage display enabled compilation of libraries of antigen-binding fragments and screening with target antigens. This review discusses the advantages and disadvantages of phage display, including use of single-domain antibody technology based on the heavy chain variable domain. We describe the state-of-the-art (and practical results of its application) technology enabling production of human antibodies by sorting and sequencing the genome of individual memory B cells, using monoclonal virus-neutralizing antibodies against SARS-CoV-2 as an example. The prospects of further development of the recombinant human antibody production technology are discussed;in particular, we consider creation of sequences of variable fragments of antibodies with the help of artificial intelligence. © 2022 Group of Companies Med Expert, LLC. All rights reserved.

7.
Bulletin of Russian State Medical University ; 2023(1):12-20, 2023.
Article in English | EMBASE | ID: covidwho-2285740

ABSTRACT

Several COVID-19 vaccines have been developed in the last three years using various tecnhiques. Multiple virus-neutralizing antibodies against SARS-CoV-2 have been also obtained to combat the pandemic. However, the use of these medications for prevention and potential treatment faces significant challenges due to the emergence of new mutant virus variants, both more contagious and escaping neutralization by the immune system, that is why it is necessary to continuously renew the vaccines and develop new therapeutic antibodies. The study was aimed to use the technology of generating single-domain antibodies (nanobodies) to target the surface spike (S) protein RBD conserved epitope of the broad spectrum of SARS-CoV-2 variants. Recombinant proteins that corresponded to RBDs of three important SARS-SoV-2 strains and the full-length S protein (Wuhan) were used as antigens for immunization of a camel in order to induce production of appropriate antibodies and/or as immobilized proteins for further cross selection of the nanobody clones with pre-set specificity by the phage display. A nanobody capable of effectively recognizing the conservative region in the S protein RBDs of the broad spectrum of pandemic SARS-CoV-2 variants, including Omicron, was selected from the generated immune library. Along with conventional use in immunoassays and diagnosis, the generated nanobody can be potentially used as a module for target-specific binding used to trap coronavirus in human upper airways during the development of novel combination antiviral drugs.Copyright © 2023 Pirogov Russian National Research Medical University. All rights reserved.

8.
Vaccines (Basel) ; 11(2)2023 Feb 06.
Article in English | MEDLINE | ID: covidwho-2262529

ABSTRACT

Most neutralizing antibodies neutralize the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by directly blocking the interactions between the spike glycoprotein receptor-binding domain (RBD) and its receptor, human angiotensin-converting enzyme 2 (ACE2). Here, we report a novel nanobody (Nb) identified by an RBD-ACE2 competitive panning method that could specifically bind to the RBD of SARS-CoV-2 with a high affinity (EC50 = 0.03 nM) and successfully block the binding between the RBD and ACE2 recombinant protein. A structural simulation of the RBD-VHH complex also supports a mechanism of the Nb to block the interaction between the RBD and ACE2. A pseudovirus assay of the Nb showed it could neutralize the WT pseudovirus with high potency (IC50 = 0.026 µg/mL). Furthermore, we measured its binding to phages displaying RBDs of different SARS-CoV-2 variants and found that it could bind to recombinant phages displaying the RBD of beta and delta variants. This study also provides a method of phage library competitive panning, which could be useful for directly screening high-affinity antibodies targeting important functional regions.

9.
Int J Mol Sci ; 24(5)2023 Feb 27.
Article in English | MEDLINE | ID: covidwho-2261740

ABSTRACT

We recently reported the isolation and characterization of anti-SARS-CoV-2 antibodies from a phage display library built with the VH repertoire of a convalescent COVID-19 patient, paired with four naïve synthetic VL libraries. One of the antibodies, called IgG-A7, neutralized the Wuhan, Delta (B.1.617.2) and Omicron (B.1.1.529) strains in authentic neutralization tests (PRNT). It also protected 100% transgenic mice expressing the human angiotensin-converting enzyme 2 (hACE-2) from SARS-CoV-2 infection. In this study, the four synthetic VL libraries were combined with the semi-synthetic VH repertoire of ALTHEA Gold Libraries™ to generate a set of fully naïve, general-purpose, libraries called ALTHEA Gold Plus Libraries™. Three out of 24 specific clones for the RBD isolated from the libraries, with affinity in the low nanomolar range and sub-optimal in vitro neutralization in PRNT, were affinity optimized via a method called "Rapid Affinity Maturation" (RAM). The final molecules reached sub-nanomolar neutralization potency, slightly superior to IgG-A7, while the developability profile over the parental molecules was improved. These results demonstrate that general-purpose libraries are a valuable source of potent neutralizing antibodies. Importantly, since general-purpose libraries are "ready-to-use", it could expedite isolation of antibodies for rapidly evolving viruses such as SARS-CoV-2.


Subject(s)
COVID-19 , Animals , Humans , Mice , Antibodies, Neutralizing , Antibodies, Viral , Immunoglobulin G , Mice, Transgenic , SARS-CoV-2
10.
J Biomed Sci ; 29(1): 108, 2022 Dec 22.
Article in English | MEDLINE | ID: covidwho-2266799

ABSTRACT

BACKGROUND: The variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) harbor diverse spike (S) protein sequences, which can greatly influence the efficacies of therapeutics. Therefore, it would be of great value to develop neutralizing monoclonal antibodies (mAbs) that can broadly recognize multiple variants. METHODS: Using an mRNA-LNP immunization strategy, we generated several mAbs that specifically target the conserved S2 subunit of SARS-CoV-2 (B-S2-mAbs). These mAbs were assessed for their neutralizing activity with pseudotyped viruses and binding ability for SARS-CoV-2 variants. RESULTS: Among these mAbs, five exhibited strong neutralizing ability toward the Gamma variant and also recognized viral S proteins from the Wuhan, Alpha, Beta, Gamma, Delta and Omicron (BA.1, BA.2 and BA.5) variants. Furthermore, we demonstrated the broad reactivities of these B-S2-mAbs in several different applications, including immunosorbent, immunofluorescence and immunoblotting assays. In particular, B-S2-mAb-2 exhibited potent neutralization of Gamma variant (IC50 = 0.048 µg/ml) in a pseudovirus neutralization assay. The neutralizing epitope of B-S2-mAb-2 was identified by phage display as amino acid residues 1146-1152 (DSFKEEL) in the S2 subunit HR2 domain of SARS-CoV-2. CONCLUSION: Since there are not many mAbs that can bind the S2 subunit of SARS-CoV-2 variants, our set of B-S2-mAbs may provide important materials for basic research and potential clinical applications. Importantly, our study results demonstrate that the viral S2 subunit can be targeted for the production of cross-reactive antibodies, which may be used for coronavirus detection and neutralization.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Antibodies, Viral , Antibodies, Monoclonal/metabolism , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Neutralizing
11.
Antiviral Res ; 212: 105576, 2023 04.
Article in English | MEDLINE | ID: covidwho-2266454

ABSTRACT

Rapid emergence of new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has prompted an urgent need for the development of broadly applicable and potently neutralizing antibody platform against the SARS-CoV-2, which can be used for combatting the coronavirus disease 2019 (COVID-19). In this study, based on a noncompeting pair of phage display-derived human monoclonal antibodies (mAbs) specific to the receptor-binding domain (RBD) of SARS-CoV-2 isolated from human synthetic antibody library, we generated K202.B, a novel engineered bispecific antibody with an immunoglobulin G4-single-chain variable fragment design, with sub- or low nanomolar antigen-binding avidity. Compared with the parental mAbs or mAb cocktail, the K202.B antibody showed superior neutralizing potential against a variety of SARS-CoV-2 variants in vitro. Furthermore, structural analysis of bispecific antibody-antigen complexes using cryo-electron microscopy revealed the mode of action of K202.B complexed with a fully open three-RBD-up conformation of SARS-CoV-2 trimeric spike proteins by simultaneously interconnecting two independent epitopes of the SARS-CoV-2 RBD via inter-protomer interactions. Intravenous monotherapy using K202.B exhibited potent neutralizing activity in SARS-CoV-2 wild-type- and B.1.617.2 variant-infected mouse models, without significant toxicity in vivo. The results indicate that this novel approach of development of immunoglobulin G4-based bispecific antibody from an established human recombinant antibody library is likely to be an effective strategy for the rapid development of bispecific antibodies, and timely management against fast-evolving SARS-CoV-2 variants.


Subject(s)
Antibodies, Bispecific , COVID-19 , Animals , Mice , Humans , SARS-CoV-2/metabolism , Antibodies, Viral , Antibodies, Bispecific/pharmacology , Cryoelectron Microscopy , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus
12.
Int J Mol Sci ; 24(4)2023 Feb 06.
Article in English | MEDLINE | ID: covidwho-2237101

ABSTRACT

The rapid identification and recognition of COVID-19 have been challenging since its outbreak. Multiple methods were developed to realize fast monitoring early to prevent and control the pandemic. In addition, it is difficult and unrealistic to apply the actual virus to study and research because of the highly infectious and pathogenic SARS-CoV-2. In this study, the virus-like models were designed and produced to replace the original virus as bio-threats. Three-dimensional excitation-emission matrix fluorescence and Raman spectroscopy were employed for differentiation and recognition among the produced bio-threats and other viruses, proteins, and bacteria. Combined with PCA and LDA analysis, the identification of the models for SARS-CoV-2 was achieved, reaching a correction of 88.9% and 96.3% after cross-validation, respectively. This idea might provide a possible pattern for detecting and controlling SARS-CoV-2 from the perspective of combining optics and algorithms, which could be applied in the early-warning system against COVID-19 or other bio-threats in the future.


Subject(s)
Bacteriophages , COVID-19 , Humans , SARS-CoV-2 , Disease Outbreaks
13.
Int J Mol Sci ; 24(2)2023 Jan 12.
Article in English | MEDLINE | ID: covidwho-2236480

ABSTRACT

Phages are highly ubiquitous biological agents, which means they are ideal tools for molecular biology and recombinant DNA technology. The development of a phage display technology was a turning point in the design of phage-based vaccines. Phages are now recognized as universal adjuvant-free nanovaccine platforms. Phages are well-suited for vaccine design owing to their high stability in harsh conditions and simple and inexpensive large-scale production. The aim of this review is to summarize the overall breadth of the antiviral therapeutic perspective of phages contributing to the development of phage-based vaccines for COVID-19. We show that phage vaccines induce a strong and specific humoral response by targeted phage particles carrying the epitopes of SARS-CoV-2. Further, the engineering of the T4 bacteriophage by CRISPR (clustered regularly interspaced short palindromic repeats) presents phage vaccines as a valuable platform with potential capabilities of genetic plasticity, intrinsic immunogenicity, and stability.


Subject(s)
Bacteriophages , COVID-19 , Vaccines , Humans , Bacteriophages/genetics , COVID-19 Vaccines/genetics , COVID-19/therapy , COVID-19/genetics , SARS-CoV-2/genetics , Bacteriophage T4/genetics , Clustered Regularly Interspaced Short Palindromic Repeats
14.
Protein Sci ; 32(3): e4575, 2023 03.
Article in English | MEDLINE | ID: covidwho-2209205

ABSTRACT

The newly emerged SARS-CoV-2 causing coronavirus disease (COVID-19) resulted in >500 million infections. A great deal about the molecular processes of virus infection in the host is getting uncovered. Two sequential proteolytic cleavages of viral spike protein by host proteases are prerequisites for the entry of the virus into the host cell. The first cleavage occurs at S1/S2 site by the furin protease, and the second cleavage at a fusion activation site, the S2' site, by the TMPRSS2 protease. S2' cleavage site is present in the S2 domain of spike protein followed by a fusion peptide. Given the S2' site to be conserved among all the SARS-CoV-2 variants, we chose an S2' epitope encompassing the S2' cleavage site and generated single-chain antibodies (scFvs) through an exhaustive phage display library screening. Crystal structure of a scFv in complex with S2' epitope was determined. Incidentally, S2' epitope in the scFv bound structure adopts an alpha-helical conformation equivalent to the conformation of the epitope in the spike protein. Furthermore, these scFvs can bind to the spike protein expressed either in vitro or on the mammalian cell surface. We illustrate a molecular model based on structural and biochemical insights into the antibody-S2' epitope interaction emphasizing scFvs mediated blocking of virus entry into the host cell by restricting the access of TMPRSS2 protease and consequently inhibiting the S2' cleavage competitively.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , SARS-CoV-2/metabolism , Epitopes , Membrane Fusion , Spike Glycoprotein, Coronavirus/chemistry , Peptide Hydrolases , Virus Internalization , Mammals
15.
Romanian Archives of Microbiology and Immunology ; 81:65-67, 2022.
Article in English | ProQuest Central | ID: covidwho-2169459

ABSTRACT

Anticorpii joacă un rol esenţial în imunitatea antiinfecţioasă, fiind principalele biomolecule instrumentale pentru prevenirea sau modularea specifică a infecţiilor virale. Obiective. Studiul de faţă a urmărit actualizarea informaţiilor ştiinţifice privind anticorpii cu potenţial imunoprofilactic şi terapeutic în viroze de importanţă majoră pentru sănătatea publică (rabie, arboviroze, COVID-19, gripa cu tulpini înalt patogene), precum şi tehnologiile contemporane de obţinere a anticorpilor în laborator, pentru a fitestaţi şi utilizaţi în clinică. Materiale şi metode. Au fost studiate datele din literatura ştiinţifică de specialitate publicate în ultimii 10 ani, cu deosebire cele cu impact ştiinţific şi profesional de excelenţă (grupurile editoriale Nature, Science, Lancet, Elsevier, MDPI etc). Informaţiile au fost filtrate prin experienţa practică de lucru în domeniu a autorului, la nivel naţional şi internaţional. Rezultate şi discuţii. În prezent, pentru a genera anticorpi monoclonali, există o paletă largă de tehnologii care se utilizează în laborator: a) fuzionarea limfocitelor B cu linii de celule de mielom uman sau uman/murin;b) imortalizarea limfocitelor B prin transformare cu virusul Epstein-Barr;c) tehnologia denumită phage display care implică exprimarea regiunilor variabile (VH şi VL) ale imunoglobulinei G pe suprafaţa unor bacteriofagi, care sunt apoi selectaţi iterativ prin legarea la un antigen specific;d) tehnicile de clonare celulară prin sortarea celulelor B de memorie specifice pentru un antigen, cultivarea acestor in vitro cu activare antigen specifică, urmată de amplificarea genică şi clonarea regiunilor variabile ale lanţurilor grele şi uşoare, pornind de la o singură celulă B;ulterior regiunile clonate se exprimă în vectori specifici de expresie proteică. e) tehnologii bazate pe ADN şi ARN mesager. Pe de altă parte, se pot obţine in vitro anticorpi monoclonali, himerici, bispecifici, fragmente de anticorpi, cu proprietăţi funcţionale şi farmacologice particulare. Până la sfarşitul anului 2021 existau aproximativ 100 de tipuri de anticorpi aprobaţi pentru utilizări în clinica infecţiilor virale, la nivel mondial. Concluzii. Îmbunătăţirea şi apariţia noi abordări ale tehnologiilor de clonare celulară şi moleculară, precum şi industrializarea proceselor în condiţii automatizate, conduc la posibilitatea de a dezvolta anticorpi monoclonali cu înaltă eficienţă şi scurtarea considerabilă a timpului necesar de control şi evaluare a acestora pentru înregistrarea punerii pe piaţă. Utilizarea anticorpilor neutralizanţi rămâne una dintre cele mai promiţătoare opţiuni în lupta împotriva infecţiilor virale, fiind o decizie strategică în contextul apariţiei de noi epidemii sau al ameninţărilor bioteroriste. Sursa de finanţare: lucrări desfăşurate în cadrul proiectului PSCD/2022 - VIROMAB H, finanţat de Ministerul Apărării Naţionale.Alternate :Antibodies play an essential role in anti-infective immunity, being the main instrumental biomolecules for the specific prevention or modulation of viral infections. Objectives. The present study aimed to update scientific information regarding antibodies with immunoprophylactic and therapeutic potential in viroses of major importance for public health (rabies, arboviruses, COVID-19, influenza with highly pathogenic strains), as well as contemporary technologies for getting antibodies in the laboratory, in order to be tested and used in the clinic. Materials and methods. Data from specialized scientific literature published in the last 10 years were studied, especially those publications with excellent scientific and professional impact (publishing groups Nature, Science, Lancet, Elsevier, MDPI etc.). The information has been filtered through the author's practical work experience in the field, at national and international level. Results and discussion. Currently, to generate monoclonal antibodies, there is a wide range of te hnologies that are used in the laboratory: a) fusion of B lymphocytes with human or human/murine myeloma cell lines;b) immortalization of B lymphocytes by transformation with the Epstein-Barr virus;c) the phage display technology that involves the expression of the variable regions (VH and VL) of immunoglobulin G on the surface of some bacteriophages, which are then iteratively selected by binding to a specific antigen;d) cell cloning techniques by sorting memory B cells specific for an antigen, in vitro cell cultivation with specific antigen activation, followed by gene amplification and cloning of the variable regions of the heavy and light chains (VH, VL) starting from a single B cell;subsequently the cloned regions are expressed in specific vectors. e) technologies based on DNA and messenger RNA. On the other hand, monoclonal, chimeric, bispecific antibodies, antibody fragments can be obtained in vitro, with particular functional and pharmacological properties. By the end of 2021 there were approximately 100 types of antibodies approved for use in the clinic of viral infections, worldwide. Conclusions. The improvement and the emergence of new approaches to cellular and molecular cloning technologies, as well as the industrialization of processes under automated conditions, lead to the possibility of developing monoclonal antibodies with high efficiency and the considerable shortening of the time required for their control and evaluation for market registration. The use of neutralizing antibodies remains one of the most promising options in the fight against viral infections, being a strategic decision in the context of the emergence of new epidemics or bioterrorist threats. Funding source: works carried out within the project PSCD/2022 - VIROMAB H, funded by the Ministry of National Defense.

16.
Viruses ; 15(1)2023 Jan 06.
Article in English | MEDLINE | ID: covidwho-2166972

ABSTRACT

The rapid emergence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has resulted in the ongoing global coronavirus disease 2019 (COVID-19) pandemic. Thus, the rapid development of a platform to detect a broad range of SARS-CoV-2 variants is essential for successful COVID-19 management. In this study, four SARS-CoV-2 spike protein-specific single-chain variable fragments (scFvs) were isolated from a synthetic antibody library using phage display technology. Following the conversion of these scFvs into monoclonal antibodies (mAbs) (K104.1-K104.4) and production and purification of the mAbs, the antibody pair (K104.1 and K104.2) that exhibited the highest binding affinity (K104.1 and K104.2, 1.3 nM and 1.9 nM) was selected. Biochemical analyses revealed that this antibody pair specifically bound to different sites on the S2 subunit of the spike protein. Furthermore, we developed a highly sensitive sandwich immunoassay using this antibody pair that accurately and quantitatively detected the spike proteins of wild-type SARS-CoV-2 and multiple variants, including Alpha, Beta, Gamma, Delta, Kappa, and Omicron, in the picomolar range. Conclusively, the novel phage display-derived mAbs we have developed may be useful for the rapid and efficient detection of the fast-evolving SARS-CoV-2.


Subject(s)
Antibodies, Monoclonal , Bacteriophages , SARS-CoV-2 , Single-Chain Antibodies , Humans , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , SARS-CoV-2/genetics , Single-Chain Antibodies/genetics , Spike Glycoprotein, Coronavirus
17.
Basic Clin Androl ; 32(1): 22, 2022 Nov 17.
Article in English | MEDLINE | ID: covidwho-2139142

ABSTRACT

BACKGROUND: At present, there is a lack of standardized preparation methods of sperm antigen for the detection of antisperm antibody (AsAb). To screen sperm antigen mimotopes from a phage display random peptide library and use them to establish an enzyme-linked immunosorbent assay (ELISA) for the detection of AsAb, immunoglobulins were extracted from the sera of rabbits with positive AsAb and negative AsAb, respectively, by the saturated ammonium sulfate method, and a phage display 12-mer peptide library was affinity panned by the extracted immunoglobins coated on the ELISA plate. Then, the obtained positive phage clones were identified by ELISA and sent for sequencing and peptides synthesis. Last, a diagnostic ELISA was established to detect clinical serum and seminal plasma samples. RESULTS: A total of sixty phage clones were chosen by affinity panning, and sixteen of them reacted positively with AsAb in indirect ELISA and sandwich ELISA. Following DNA sequencing and translation, the peptide sequences of the sixteen positive clones were obtained. By comparison in Blast database, four of sixteen positive clones were found to be closely related to male reproduction. Two (#1 and #25) of four mimotopes were synthesized, and an ELISA method was established using the two mimotopes as sperm specific antigens. One hundred and thirty-four serum samples and seventy-four seminal plasma samples from infertile couples were analyzed by the established ELISA with #1 and #25 mimotopes, respectively. The positive rates of AsAb in serum samples were 20.15% (27/134) for #1 and 11.19% (15/134) for #25, respectively, and the coincidence rate between them was 91.04% (122/134). The positive rates of AsAb in seminal plasma samples were 1.35% (1/74) for both #1 and #25, and the coincidence rate was 100%. CONCLUSION: Sperm antigen mimotopes can be obtained successfully by the phage display technique, and can be used as standard sperm specific antigens to establish an ELISA method for the detection of AsAb.


RéSUMé: CONTEXTE: À ce jour, il n'existe pas de méthodes normalisées de préparation d'antigènes spermatiques pour la détection des anticorps anti-spermatozoïdes (ACAS). Dans le but d'élaborer un tel test ELISA (enzyme-linked immunosorbent assay), nous avons extrait de sérum de lapins des anticorps anti-spermatozoïdes humains via la technique du sulfate d'ammonium saturé et en ayant recours à une librairie phagique de peptides (12-mer). Les clones positifs ont été identifiés par ELISA, séquencés à façon et les peptides correspondants ont été synthétisés. In fine, un test ELISA diagnostic a été conçu pour être utilisé avec des échantillons cliniques de sérum et de plasmas séminaux. RéSULTATS: Au total, soixante clones de phages ont été sélectionnés, et seize d'entre eux se sont avérés interagir avec les ACAS en ELISA indirect comme en ELISA sandwich. Les séquences peptidiques de ces seize clones positifs ont été obtenues. Par comparaison avec les bases de données (Blast), quatre de ces seize clones positifs se sont révélés être étroitement liés à la reproduction masculine. Deux des quatre mimotopes (#1 et #25) ont été synthétisés, et un test ELISA a été généré en utilisant ces deux mimotopes comme antigènes spécifiques des spermatozoïdes. Cent trente-quatre échantillons de sérum et soixante-quatorze échantillons de plasma séminal de patients de couples infertiles ont alors été analysés avec ce test ELISA. Respectivement, les échantillons sériques se sont révélés positifs à 20,15% (27/134) pour le mimotope #1 et à 11,19% (15/134) pour le mimotope #25, avec un taux de coïncidence de 91,04% (122/134). Seul un échantillon de plasma séminal (1/74, soit 1, 35%) s'est révélé positif à la fois pour le mimotope #1 et #25 (coïncidence 100%). CONCLUSION: La technique « phage display¼ nous a permis d'identifier des mimotopes d'antigènes spermatiques qui ont pu être utilisés afin de générer un test ELISA pour la détection d'anticorps anti-spermatozoïdes.

18.
FEBS Open Bio ; 12(11): 2057-2064, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2093221

ABSTRACT

Proteinaceous protease inhibitors can strongly and specifically inhibit cognate proteases, but their use as pharmaceuticals is limited by their size. As such, the development of effective protease peptide inhibitors would be beneficial for biochemical studies and drug discovery. In this study, we applied a phage display system to select subtilisin BPN'-binding peptides and evaluated their inhibitory activities against subtilisin BPN'. A 12mer peptide with an intramolecular disulfide bond inhibited subtilisin BPN' (Ki value of 13.0 nm). Further mutational analyses of the peptide resulted in the development of a short peptide inhibitor against subtilisin BPN' that showed high inhibitory activity and binding affinity (Ki value of 0.30 nm). This activity was found to be derived from the conformational rigidity caused by the intramolecular disulfide bond and the small residue at the P1' site and from the interaction of the P4 and P6' residues with subtilisin BPN'.


Subject(s)
Peptides , Subtilisins , Subtilisins/chemistry , Subtilisins/metabolism , Mutagenesis, Site-Directed , Peptides/pharmacology , Protease Inhibitors , Disulfides
19.
Cells ; 11(21)2022 Oct 24.
Article in English | MEDLINE | ID: covidwho-2082270

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an infectious disease that has become a serious burden on global public health. This study screened and yielded specific nanobodies (Nbs) against SARS-CoV-2 spike protein receptor binding domain (RBD), following testing its basic characteristics. A nanobody phage library was established by immunizing a camel with RBD protein. After three rounds of panning, the positive colonies were screened by enzyme-linked immunosorbent assay (ELISA). By sequencing, four different sequences of nanobody gene fragments were selected. The four nanobody fusion proteins were expressed and purified, respectively. The specificity and affinity of the four nanobodies were identified by ELISA. Our results showed that an immune phage display library against SARS-CoV-2 has been successfully constructed with a library capacity of which was 4.7 × 108 CFU. The four purified nanobodies showed specific high-affinity binding SARS-CoV-2 S-RBD. Among these, the antigen binding affinity of Nb61 was more comparable to that of commercial rabbit anti-SARS-CoV-2 S-RBD antibodies. In sum, our study has obtained four nanobody strains against SARS-CoV-2 S-RBD with significant affinity and specificity, therefore laying an essential foundation for further research as well as the applications of diagnostic and therapeutic tools of SARS-CoV-2.


Subject(s)
COVID-19 , Single-Domain Antibodies , Animals , Humans , Rabbits , Spike Glycoprotein, Coronavirus/chemistry , Antibodies, Neutralizing , SARS-CoV-2 , Camelus
20.
Sens Actuators B Chem ; 374: 132780, 2023 Jan 01.
Article in English | MEDLINE | ID: covidwho-2069694

ABSTRACT

In this work, the nucleic acid detection of SARS-Cov-2 is extended to protein markers of the virus, utilizing bacteriophage. Specifically, the phage display technique enables the main protease of SARS-Cov-2 to control the self-replication of m13 phage, so that the presence of the viral protease can be amplified by phage replication as the first round of signal amplification. Then, the genome of replicated phage can be detected using polymer chain reaction (PCR), as the second round of signal amplification. Based on these two types of well-established biotechnology, the proposed method shows satisfactory sensitivity and robustness in the direct serum detection of the viral protease. These results may point to clinical application in the near future.

SELECTION OF CITATIONS
SEARCH DETAIL