Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 6935, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138197

ABSTRACT

Nanobodies (Nbs), the smallest antigen-binding fragments with high stability and affinity derived from the variable domain of naturally occurring heavy-chain-only antibodies in camelids, have been shown as an efficient way to improve the specificity to tumors for photodynamic therapy (PDT). Nonetheless, the rapid clearance of Nbs in vivo restricts the accumulation and retention of the photosensitizer at the tumor site causing insufficient therapeutic outcome, especially in large-volume tumors. Herein, we develop photodynamic conjugates, MNB-Pyra Nbs, through site-specific conjugation between 7D12 Nbs and type I photosensitizer MNB-Pyra (morpholine-modified nile blue structure connected to pyrazolinone) in a 1:2 ratio. The photosensitizers with long-term retention can be released at the tumor site by reactive oxygen species cleavage after illumination, accompanied with fluorescence recovery for self-reporting the occurrence of PDT. Ultimately, a single dose of MNB-Pyra Nbs demonstrate highly effective tumor suppression with high biosafety in the large-volume tumor models after three rounds of PDT. This nanobody conjugate provides a paradigm for the design of precise long-time retention photosensitizers and is expected to promote the development of PDT.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Animals , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/chemistry , Mice , Humans , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Mice, Inbred BALB C , Female , Neoplasms/drug therapy , Neoplasms/immunology , Mice, Nude , Xenograft Model Antitumor Assays
2.
Sci Rep ; 14(1): 19074, 2024 08 17.
Article in English | MEDLINE | ID: mdl-39154093

ABSTRACT

Single-domain antibodies (sdAbs) or nanobodies have received widespread attention due to their small size (~ 15 kDa) and diverse applications in bio-derived therapeutics. As many modern biotechnology breakthroughs are applied to antibody engineering and design, nanobody thermostability or melting temperature (Tm) is crucial for their successful utilization. In this study, we present TEMPRO which is a predictive modeling approach for estimating the Tm of nanobodies using computational methods. Our methodology integrates various nanobody biophysical features to include Evolutionary Scale Modeling (ESM) embeddings, NetSurfP3 structural predictions, pLDDT scores per sdAb region from AlphaFold2, and each sequence's physicochemical characteristics. This approach is validated with our combined dataset containing 567 unique sequences with corresponding experimental Tm values from a manually curated internal data and a recently published nanobody database, NbThermo. Our results indicate the efficacy of protein embeddings in reliably predicting the Tm of sdAbs with mean absolute error (MAE) of 4.03 °C and root mean squared error (RMSE) of 5.66 °C, thus offering a valuable tool for the optimization of nanobodies for various biomedical and therapeutic applications. Moreover, we have validated the models' performance using experimentally determined Tms from nanobodies not found in NbThermo. This predictive model not only enhances nanobody thermostability prediction, but also provides a useful perspective of using embeddings as a tool for facilitating a broader applicability of downstream protein analyses.


Subject(s)
Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Transition Temperature , Models, Molecular , Protein Stability , Computational Biology/methods
3.
PLoS Pathog ; 20(8): e1012438, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39141662

ABSTRACT

The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe and fatal acute respiratory disease in humans. High fatality rates and continued infectiousness remain a pressing concern for global health preparedness. Antibodies targeted at the receptor-binding domain (RBD) are major countermeasures against human viral infection. Here, we report four potent nanobodies against MERS-CoV, which are isolated from alpaca, and especially the potency of Nb14 is highest in the pseudotyped virus assay. Structural studies show that Nb14 framework regions (FRs) are mainly involved in interactions targeting a novel epitope, which is entirely distinct from all previously reported antibodies, and disrupt the protein-carbohydrate interaction between residue W535 of RBD and hDPP4 N229-linked carbohydrate moiety (hDPP4-N229-glycan). Different from Nb14, Nb9 targets the cryptic face of RBD, which is distinctive from the hDPP4 binding site and the Nb14 epitope, and it induces the ß5-ß6 loop to inflect towards a shallow groove of the RBD and dampens the accommodation of a short helix of hDPP4. The particularly striking epitopes endow the two Nbs administrate synergistically in the pseudotyped MERS-CoV assays. These results not only character unprecedented epitopes for antibody recognition but also provide promising agents for prophylaxis and therapy of MERS-CoV infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Epitopes , Middle East Respiratory Syndrome Coronavirus , Single-Domain Antibodies , Middle East Respiratory Syndrome Coronavirus/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Humans , Epitopes/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Animals , Coronavirus Infections/immunology , Coronavirus Infections/virology , Camelids, New World/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Mice , Receptors, Virus/metabolism , Receptors, Virus/immunology
4.
Biochem Pharmacol ; 227: 116457, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098732

ABSTRACT

The chemokine receptor CXCR4 is involved in the development and migration of stem and immune cells but is also implicated in tumor progression and metastasis for a variety of cancers. Antagonizing ligand (CXCL12)-induced CXCR4 signaling is, therefore, of therapeutic interest. Currently, there are two small-molecule CXCR4 antagonists on the market for the mobilization of hematopoietic stem cells. Other molecules with improved potencies and safety profiles are being developed for different indications, including cancer. Moreover, multiple antagonistic nanobodies targeting CXCR4 displayed similar or better potencies as compared to the CXCR4-targeting molecule AMD3100 (Plerixafor), which was further enhanced through avid binding of bivalent derivatives. In this study, we aimed to compare the affinities of various multivalent nanobody formats which might be differently impacted by avidity. By fusion to a flexible GS-linker, Fc-region of human IgG1, different C4bp/CLR multimerization domains, or via site-directed conjugation to a trivalent linker scaffold, we generated different types of multivalent nanobodies with varying valencies ranging from bivalent to decavalent. Of these, C-terminal fusion, especially to human Fc, was most advantageous with a 2-log-fold and 3-log-fold increased potency in inhibiting CXCL12-mediated Gαi- or ß-arrestin recruitment, respectively. Overall, we describe strategies for generating multivalent and high-potency CXCR4 antagonistic nanobodies able to induce receptor clustering and conclude that fusion to an Fc-tail results in the highest avidity effect irrespective of the hinge linker.


Subject(s)
Receptors, CXCR4 , Single-Domain Antibodies , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, CXCR4/immunology , Humans , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Animals , Chemokine CXCL12/metabolism , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/immunology , HEK293 Cells , Antibody Affinity
5.
Biosens Bioelectron ; 263: 116598, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39094292

ABSTRACT

Antibody pairs-based immunoassay platforms served as essential and effective tools in the field of pathogen detection. However, the cumbersome preparation and limited detection sensitivity of antibody pairs challenge in establishment of a highly sensitive detection platform. In this study, using COVID-19 testing as a case, we utilized readily accessible nanobodies as detection antibodies and further proposed an accurate design concept with a more scientific and efficient screening strategy to obtain ultrasensitive antibody pairs. We employed nanobodies capable of binding different antigenic epitopes of the nucleocapsid (NP) or receptor-binding domain (RBD) antigens sandwich as substitutes for monoclonal antibodies (mAbs) sandwich in fast detection formats and utilized time-resolved fluorescence (TRF) microspheres as the signal probe. Consequently, we developed a multi-epitope nanobody sandwich-based fluorescence lateral flow immunoassay (FLFA) strip. Our results suggest that the NP antigen had a detection limit of 12.01pg/mL, while the RBD antigen had a limit of 6.51 pg/mL using our FLFA strip. Based on double mAb sandwiches, the values presented herein demonstrated 4 to 32-fold enhancements in sensitivity, and 32 to 256-fold enhancements compared to commercially available antigen lateral flow assay kits. Furthermore, we demonstrated the excellent characteristics of the proposed test strip, including its specificity, stability, accuracy, and repeatability, which underscores its the prospective utility. Indeed, these findings indicate that our established screening strategy along with the multi-epitope nanobody sandwich mode provides an optimized strategy in the field of pathogen detection.


Subject(s)
Biosensing Techniques , COVID-19 , SARS-CoV-2 , Single-Domain Antibodies , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , COVID-19/diagnosis , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , SARS-CoV-2/isolation & purification , Humans , Biosensing Techniques/methods , Antibodies, Viral/immunology , Antibodies, Viral/blood , Limit of Detection , Immunoassay/methods , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , COVID-19 Testing/methods , COVID-19 Serological Testing/methods , Antigens, Viral/immunology
6.
Nat Commun ; 15(1): 7228, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174501

ABSTRACT

The Wnt receptor Frizzled3 (FZD3) is important for brain axonal development and cancer progression. We report structures of FZD3 in complex with extracellular and intracellular binding nanobodies (Nb). The crystal structure of Nb8 in complex with the FZD3 cysteine-rich domain (CRD) reveals that the nanobody binds at the base of the lipid-binding groove and can compete with Wnt5a. Nb8 fused with the Dickkopf-1 C-terminal domain behaves as a FZD3-specific Wnt surrogate, activating ß-catenin signalling. The cryo-EM structure of FZD3 in complex with Nb9 reveals partially resolved density for the CRD, which exhibits positional flexibility, and a transmembrane conformation that resembles active GPCRs. Nb9 binds to the cytoplasmic region of FZD3 at the putative Dishevelled (DVL) or G protein-binding site, competes with DVL binding, and inhibits GαS coupling. In combination, our FZD3 structures with nanobody modulators map extracellular and intracellular interaction surfaces of functional, and potentially therapeutic, relevance.


Subject(s)
Frizzled Receptors , Single-Domain Antibodies , Frizzled Receptors/metabolism , Frizzled Receptors/chemistry , Humans , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Protein Binding , Crystallography, X-Ray , HEK293 Cells , Binding Sites , Cryoelectron Microscopy , Animals , Models, Molecular , Protein Domains , Dishevelled Proteins/metabolism , Dishevelled Proteins/chemistry , Dishevelled Proteins/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , beta Catenin/chemistry
7.
Nat Commun ; 15(1): 6892, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134522

ABSTRACT

Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.


Subject(s)
Antibodies, Viral , Cryoelectron Microscopy , Epitopes , Henipavirus Infections , Nipah Virus , Single-Domain Antibodies , Nipah Virus/immunology , Humans , Animals , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/virology , Epitopes/immunology , Mice , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Antibodies, Viral/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Female , HEK293 Cells
8.
Anal Chim Acta ; 1319: 342986, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39122284

ABSTRACT

BACKGROUND: Due to the serious issue of ofloxacin (OFL) abuse, there is an increasingly urgent need for accurate and rapid detection of OFL. Immunoassay has become the "golden method" for detecting OFL in complex matrix beneficial to its applicability for a large-scale screening, rapidity, and simplicity. However, traditional antibodies used in immunoassay present challenges such as time-consuming preparation, unstable sensitivity and specificity, and difficulty in directional evolution. In this paper, we successfully developed an OFL detection method based on a shark-derived single-domain antibody (ssdAb) to address these issues. RESULTS: Using phage display technology and a heterologous expression system, OFL-specific clones 1O11, 1O13, 1O17, 1O19, 1O21, and 2O26 were successfully isolated and expressed in soluble form. Among all OFL-specific ssdAbs, the 1O17 ssdAb exhibited the highest binding affinity to OFL in a concentration-dependence manner. The limit of detection (IC10) of 1O17 ssdAb was calculated as 0.34 ng/mL with a detection range of 3.40-1315.00 ng/mL, and its cross reactivity with other analogs was calculated to be less than 5.98 %, indicating high specificity and sensitivity. Molecular docking results revealed that 100Trp and 101Arg located in the CDR3 region of 1O17 ssdAb were crucial for OFL binding. In fish matrix performance tests, the 1O17 ssdAb did not demonstrate severe matrix interference in OFL-negative fish matrix, achieving satisfactory recovery rates ranging from 83.04 % to 108.82 % with high reproducibility. SIGNIFICANCE: This research provides a new and efficient OFL detection recognition element with significant potential in immunoassay applications, broadening the application scenarios of ssdAbs. It offers valuable insights into the structure-activity relationship between ssdAbs and small molecules, laying a theoretical foundation for the further directional modification and maturation of ssdAbs in subsequent applications.


Subject(s)
Ofloxacin , Sharks , Single-Domain Antibodies , Animals , Ofloxacin/analysis , Ofloxacin/immunology , Ofloxacin/chemistry , Sharks/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Limit of Detection , Immunoassay/methods
10.
Sci Adv ; 10(29): eado9413, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39018393

ABSTRACT

Chemical ligation of peptides is increasingly used to generate proteins not readily accessible by recombinant approaches. However, a robust method to ligate "difficult" peptides remains to be developed. Here, we report an enhanced native chemical ligation strategy mediated by peptide conjugation in trifluoroacetic acid (TFA). The conjugation between a carboxyl-terminal peptide thiosalicylaldehyde thioester and a 1,3-dithiol-containing peptide in TFA proceeds rapidly to form a thioacetal-linked intermediate, which is readily converted into the desired native amide bond product through simple postligation treatment. The effectiveness and practicality of the method was demonstrated by the successful synthesis of several challenging proteins, including the SARS-CoV-2 transmembrane Envelope (E) protein and nanobodies. Because of the ability of TFA to dissolve virtually all peptides and prevent the formation of unreactive peptide structures, the method is expected to open new opportunities for synthesizing all families of proteins, particularly those with aggregable or colloidal peptide segments.


Subject(s)
Peptides , Trifluoroacetic Acid , Trifluoroacetic Acid/chemistry , Peptides/chemistry , SARS-CoV-2/chemistry , Single-Domain Antibodies/chemistry , Humans , COVID-19/virology
11.
Nat Commun ; 15(1): 6214, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043660

ABSTRACT

Protein-protein interactions (PPIs) are central in cell metabolism but research tools for the structural and functional characterization of these PPIs are often missing. Here we introduce broadly applicable immunization (Cross-link PPIs and immunize llamas, ChILL) and selection strategies (Display and co-selection, DisCO) for the discovery of diverse nanobodies that either stabilize or disrupt PPIs in a single experiment. We apply ChILL and DisCO to identify competitive, connective, or fully allosteric nanobodies that inhibit or facilitate the formation of the SOS1•RAS complex and modulate the nucleotide exchange rate on this pivotal GTPase in vitro as well as RAS signalling in cellulo. One of these connective nanobodies fills a cavity that was previously identified as the binding pocket for a series of therapeutic lead compounds. The long complementarity-determining region (CDR3) that penetrates this binding pocket serves as pharmacophore for extending the repertoire of potential leads.


Subject(s)
Protein Binding , SOS1 Protein , Single-Domain Antibodies , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism , SOS1 Protein/metabolism , SOS1 Protein/chemistry , SOS1 Protein/genetics , SOS1 Protein/immunology , Humans , Animals , Allosteric Regulation , ras Proteins/metabolism , ras Proteins/chemistry , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Binding Sites , Camelids, New World/immunology , Immunization , Signal Transduction , Models, Molecular
12.
Sci Rep ; 14(1): 17747, 2024 07 31.
Article in English | MEDLINE | ID: mdl-39085444

ABSTRACT

Using conventional immunoglobulin G (IgG) molecules as therapeutic agents presents several well-known disadvantages owing to their large size and structural complexity, negatively impacting development and production efficiency. Single-domain antibodies (sdAbs) are the smallest functional antibody format (~ 15 kDa) and represent a viable alternative to IgG in many applications. However, unlike natural single-domain antibodies, such as camelid VHH, the variable domains of conventional antibodies show poor physicochemical properties when expressed as sdAbs. This report identified stable sdAb variants of human VH3-23 from a framework region 2-randomized human VH library by phage display selection under thermal challenge. Synthetic complementarity determining region diversity was introduced to one of the selected variants with high thermal stability, expression level, and monomeric content to construct a human VH sdAb library. The library was validated by panning against a panel of antigens, and target-specific binders were identified and characterized for their affinity and biophysical properties. The results of this study suggest that a synthetic sdAb library based on a stability-engineered human VH scaffold could be a facile source of high-quality sdAb for many practical applications.


Subject(s)
Complementarity Determining Regions , Peptide Library , Protein Engineering , Protein Stability , Single-Domain Antibodies , Humans , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Protein Engineering/methods , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Complementarity Determining Regions/genetics , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology
13.
Sci Rep ; 14(1): 16086, 2024 07 12.
Article in English | MEDLINE | ID: mdl-38992064

ABSTRACT

The classical amyloid cascade hypothesis postulates that the aggregation of amyloid plaques and the accumulation of intracellular hyperphosphorylated Tau tangles, together, lead to profound neuronal death. However, emerging research has demonstrated that soluble amyloid-ß oligomers (SAßOs) accumulate early, prior to amyloid plaque formation. SAßOs induce memory impairment and disrupt cognitive function independent of amyloid-ß plaques, and even in the absence of plaque formation. This work describes the development and characterization of a novel anti-SAßO (E3) nanobody generated from an alpaca immunized with SAßO. In-vitro assays and in-vivo studies using 5XFAD mice indicate that the fluorescein (FAM)-labeled E3 nanobody recognizes both SAßOs and amyloid-ß plaques. The E3 nanobody traverses across the blood-brain barrier and binds to amyloid species in the brain of 5XFAD mice. Imaging of mouse brains reveals that SAßO and amyloid-ß plaques are not only different in size, shape, and morphology, but also have a distinct spatial distribution in the brain. SAßOs are associated with neurons, while amyloid plaques reside in the extracellular matrix. The results of this study demonstrate that the SAßO nanobody can serve as a diagnostic agent with potential theragnostic applications in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Plaque, Amyloid , Single-Domain Antibodies , Animals , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Mice , Plaque, Amyloid/metabolism , Alzheimer Disease/metabolism , Humans , Brain/metabolism , Brain/pathology , Blood-Brain Barrier/metabolism , Mice, Transgenic , Camelids, New World , Disease Models, Animal
14.
Bioconjug Chem ; 35(8): 1172-1181, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-38994647

ABSTRACT

Enzymatic site-specific bioconjugation techniques, in particular sortase-mediated ligation, are increasingly used to generate conjugated proteins for a wide array of applications. Extension of the utility and practicality of sortagging for diverse purposes is critically dependent on further improvement of the efficiency of sortagging reactions with a wider structural variety of substrates. We present a comprehensive comparative mass spectrometry screening study of synthetic nonpeptidic incoming amine nucleophile substrates of Staphylococcus aureus Sortase A enzyme. We have identified the optimal structural motifs among the chemically diverse set of 452 model primary and secondary amine-containing sortagging substrates, and we demonstrate the utility of representative amine linkers for efficient C-terminal biotinylation of nanobodies.


Subject(s)
Amines , Aminoacyltransferases , Bacterial Proteins , Cysteine Endopeptidases , Staphylococcus aureus , Aminoacyltransferases/metabolism , Amines/chemistry , Staphylococcus aureus/enzymology , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/chemistry , Bacterial Proteins/chemistry , Biotinylation , Substrate Specificity , Single-Domain Antibodies/chemistry , Mass Spectrometry
15.
J Med Chem ; 67(15): 12855-12867, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39077778

ABSTRACT

The overexpression of HER2 is pivotal in the initiation and progression of breast cancer. Developing HER2-targeted radiotracers is crucial for noninvasive assessment of HER2 expression, patient selection for HER2-targeted therapy, monitoring treatment response, and identifying resistance. Here, we reported a nonsite-specific coupled radiotracer, 68Ga-NOTA-SNA004-His6, and a site-specific coupled radiotracer, 68Ga-NODAGA-SNA004-GSC, based on a novel HER2 nanobody, SNA004. Both radiotracers exhibited high affinity, specific targeting, and rapid clearance in vitro and in vivo. Additionally, these tracers and trastuzumab showed noncompetitive binding to HER2. Compared to 68Ga-NOTA-SNA004-His6, 68Ga-NODAGA-SNA004-GSC demonstrated significantly reduced renal and liver uptake. PET/CT imaging with 68Ga-NODAGA-SNA004-GSC sensitively detected the responsiveness of various tumor models to trastuzumab and its antibody-drug conjugates (ADCs). Overall, the site-specific coupled radiotracer 68Ga-NODAGA-SNA004-GSC offered significant advantages in biodistribution and signal-to-noise ratio, making it a valuable tool for monitoring HER2 expression levels before, during, and after trastuzumab and ADC treatment.


Subject(s)
Gallium Radioisotopes , Heterocyclic Compounds, 1-Ring , Immunoconjugates , Receptor, ErbB-2 , Single-Domain Antibodies , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Animals , Gallium Radioisotopes/chemistry , Female , Heterocyclic Compounds, 1-Ring/chemistry , Single-Domain Antibodies/chemistry , Mice , Tissue Distribution , Cell Line, Tumor , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Acetates/chemistry , Acetates/pharmacology , Radiopharmaceuticals/chemistry , Mice, Nude , Positron Emission Tomography Computed Tomography/methods
16.
J Hazard Mater ; 477: 135264, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39032175

ABSTRACT

Nontoxic substitutes to mycotoxins can facilitate the development of eco-friendly immunoassays. To explore a novel nontoxic substitute to ochratoxin A (OTA), this study screened shark anti-idiotypic variable new antigen receptors (VNARs) against the alpaca anti-OTA nanobody Nb28 through phage display. After four rounds of biopanning of a naïve VNAR phage display library derived from six adult Chiloscyllium plagiosum sharks, one positive clone, namely, P-3, was validated through a phage enzyme-linked immunosorbent assay (phage ELISA). The recombinant anti-idiotypic VNAR AId-V3 was obtained by prokaryotic expression, and the interactions between Nb28 and AId-V3 were investigated via computer-assisted simulation. The affinity of AId-V3 for Nb28 and its heptamer Nb28-C4bpα was measured using Biacore assay. Combining Nb28-C4bpα with AId-V3, a novel direct competitive ELISA (dcELISA) was developed for OTA analysis, with a limit of detection of 0.44 ng/mL and a linear range of 1.77-32.25 ng/mL. The good selectivity, reliability, and precision of dcELISA were confirmed via cross-reaction analysis and recovery experiments. Seven commercial pepper powder samples were tested using dcELISA and validated using high-performance liquid chromatography. Overall, the shark anti-idiotypic VNAR was demonstrated as a promising nontoxic substitute to OTA, and the proposed method was confirmed as a reliable tool for detecting OTA in food.


Subject(s)
Camelids, New World , Enzyme-Linked Immunosorbent Assay , Ochratoxins , Sharks , Single-Domain Antibodies , Ochratoxins/analysis , Ochratoxins/immunology , Sharks/immunology , Animals , Camelids, New World/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Antibodies, Anti-Idiotypic/immunology , Receptors, Antigen/immunology
17.
Int J Biol Macromol ; 276(Pt 2): 133957, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39029852

ABSTRACT

Staphylococcal Enterotoxin Type B (SEB), produced by Staphylococcus aureus bacteria, is notorious for inducing severe food poisoning and toxic shock syndrome. While nanobody-based treatments hold promises for combating SEB-induced diseases, the lack of structural information between SEB and nanobodies has hindered the development of nanobody-based therapeutics. Here, we present crystal structures of SEB-Nb3, SEB-Nb6, SEB-Nb8, SEB-Nb11, and SEB-Nb20 at resolutions ranging from 1.59 Å to 2.33 Å. Crystallographic analysis revealed that Nb3, Nb8, Nb11, and Nb20 bind to SEB at the T-cell receptor (TCR) interface, while Nb6 binds at the major histocompatibility complex (MHC) interface, suggesting their potential to inhibit SEB function by disrupting interactions with TCR or MHC molecules. Molecular biological analyses confirmed the thermodynamic and kinetic parameters of Nb3, Nb5, Nb6, Nb8, Nb11, Nb15, Nb18, and Nb20 to SEB. The competitive inhibition was further confirmed by cell-based experiments demonstrating nanobody neutralization. These findings elucidate the structural basis for developing specific nanobodies to neutralize SEB threats, providing crucial insights into the underlying mechanisms and offering significant assistance for further optimization towards future therapeutic strategies.


Subject(s)
Enterotoxins , Protein Binding , Single-Domain Antibodies , Enterotoxins/chemistry , Enterotoxins/immunology , Enterotoxins/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Humans , Models, Molecular , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/chemistry , Staphylococcus aureus/immunology , Crystallography, X-Ray , Thermodynamics , Kinetics
18.
J Nanobiotechnology ; 22(1): 388, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956618

ABSTRACT

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent swine pathogen, which has caused adverse impact on the global swine industry for almost 30 years. However, due to the immune suppression caused by the virus and the genetic diversity in PRRSV, no virus-targeting broad neutralizing strategy has been successfully developed yet. Antiviral peptide and nanobody have attracted extensive attention with the ease in production and the efficacy in practice. In this study, four new fusion proteins named nanobody peptide conjugates (NPCs) were developed by combining PRRSV specific non-neutralizing nanobodies with CD163-derived peptides targeting the receptor binding domain (RBD) of PRRSV proteins. RESULTS: Four NPCs were successfully constructed using two nanobodies against PRRSV N and nsp9 individually, recombining with two antiviral peptides 4H7 or 8H2 from porcine CD163 respectively. All four NPCs demonstrated specific capability of binding to PRRSV and broad inhibitory effect against various lineages of PRRSV in a dose-dependent manner. NPCs interfere with the binding of the RBD of PRRSV proteins to CD163 in the PRRSV pre-attachment stage by CD163 epitope peptides in the assistance of Nb components. NPCs also suppress viral replication during the stage of post-attachment, and the inhibitory effects depend on the antiviral functions of Nb parts in NPCs, including the interference in long viral RNA synthesis, NF-κB and IFN-ß activation. Moreover, an interaction was predicted between aa K31 and T32 sites of neutralizing domain 4H7 of NPC-N/nsp9-4H7 and the motif 171NLRLTG176 of PRRSV GP2a. The motif 28SSS30 of neutralizing domain 8H2 of NPC-N/nsp9-8H2 could also form hydrogens to bind with the motif 152NAFLP156 of PRRSV GP3. The study provides valuable insights into the structural characteristics and potential functional implications of the RBD of PRRSV proteins. Finally, as indicated in a mouse model, NPC intranasally inoculated in vivo for 12-24 h sustains the significant neutralizing activity against PRRSV. These findings inspire the potential of NPC as a preventive measure to reduce the transmission risk in the host population against respiratory infectious agents like PRRSV. CONCLUSION: The aim of the current study was to develop a peptide based bioactive compound to neutralize various PRRSV strains. The new antiviral NPC (nanobody peptide conjugate) consists of a specific nanobody targeting the viral protein and a neutralizing CD163 epitope peptide for virus blocking and provides significant antiviral activity. The study will greatly promote the antiviral drug R&D against PRRSV and enlighten a new strategy against other viral diseases.


Subject(s)
Antibodies, Neutralizing , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Peptides , Porcine respiratory and reproductive syndrome virus , Receptors, Cell Surface , Single-Domain Antibodies , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/drug effects , Animals , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Swine , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Receptors, Cell Surface/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Antibodies, Neutralizing/immunology , Peptides/chemistry , Peptides/pharmacology , Peptides/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Mice , Virus Replication/drug effects , Cell Line
19.
J Agric Food Chem ; 72(26): 14967-14974, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38957086

ABSTRACT

Nanobodies (Nbs) serve as powerful tools in immunoassays. However, their small size and monovalent properties pose challenges for practical application. Multimerization emerges as a significant strategy to address these limitations, enhancing the utilization of nanobodies in immunoassays. Herein, we report the construction of a Salmonella-specific fenobody (Fb) through the fusion of a nanobody to ferritin, resulting in a self-assembled 24-valent nanocage-like structure. The fenobody exhibits a 35-fold increase in avidity compared to the conventional nanobody while retaining good thermostability and specificity. Leveraging this advancement, three ELISA modes were designed using Fb as the capture antibody, along with unmodified Nb422 (FbNb-ELISA), biotinylated Nb422 (FbBio-ELISA), and phage-displayed Nb422 (FbP-ELISA) as the detection antibody, respectively. Notably, the FbNb-ELISA demonstrates a detection limit (LOD) of 3.56 × 104 CFU/mL, which is 16-fold lower than that of FbBio-ELISA and similar to FbP-ELISA. Moreover, a fenobody and nanobody sandwich chemiluminescent enzyme immunoassay (FbNb-CLISA) was developed by replacing the TMB chromogenic substrate with luminal, resulting in a 12-fold reduction in the LOD. Overall, the ferritin-displayed technology represents a promising methodology for enhancing the detection performance of nanobody-based sandwich ELISAs, thereby expanding the applicability of Nbs in food detection and other fields requiring multivalent modification.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Ferritins , Salmonella , Single-Domain Antibodies , Ferritins/immunology , Ferritins/chemistry , Ferritins/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Salmonella/immunology , Salmonella/genetics , Enzyme-Linked Immunosorbent Assay/methods , Limit of Detection , Antibody Affinity , Antibodies, Bacterial/immunology , Immunoassay/methods
20.
J Virol ; 98(7): e0070724, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38953655

ABSTRACT

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.


Subject(s)
Caliciviridae Infections , Capsid Proteins , Norovirus , Single-Domain Antibodies , Norovirus/genetics , Norovirus/drug effects , Norovirus/immunology , Humans , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Capsid Proteins/immunology , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Caliciviridae Infections/therapy , Antiviral Agents/pharmacology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/chemistry , Antibodies, Viral/immunology , Cross Reactions , Capsid/metabolism , Capsid/immunology , Blood Group Antigens/metabolism , Virus Replication/drug effects , Gastroenteritis/virology , Immunoglobulin G/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology
SELECTION OF CITATIONS
SEARCH DETAIL