Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Protein Expr Purif ; 222: 106522, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38851552

ABSTRACT

OBJECTIVE: To screen and obtain specific anti-lymphocyte activation gene-3 (LAG3) nanobody sequences, purify and express recombinant anti-LAG3 nanobody, and verify its effect on promoting T cells to kill tumor cells. METHODS: Based on the camel derived natural nanobody phage display library constructed by the research group, the biotinylated LAG3 antigen was used as the target, and the anti-LAG3 nanobody sequences were screened by biotin-streptavidin liquid phase screening, phage-ELISA and sequencing. The sequence-conjµgated human IgG1 Fc fragment was obtained, the recombinant anti-LAG3 nanobody expression vector was constructed, the expression of the recombinant anti-LAG3 nanobody was induced by IPTG and purified, and the characteristics and functions of the recombinant anti-LAG3 nanobody were verified by SDS-PAGE, Western blot, cytotoxicity assay, etc. RESULTS: One anti-LAG3 nanobody sequence was successfully screened, and the corresponding recombinant anti-LAG3 nanobody-expressing bacteria were constructed. The results of SDS-PAGE, Western blot and cytotoxicity assay showed that the recombinant anti-LAG3 nanobody was successfully expressed, which was specific, and it could promote the killing ability of T cells against tumor cells, and the optimal concentration was 200 µg/mL. CONCLUSION: The recombinant anti-LAG3 nanobody screened and expressed has specific and auxiliary anti-tumor cell effects, which lays a foundation for its subsequent application.


Subject(s)
Lymphocyte Activation Gene 3 Protein , Single-Domain Antibodies , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/pharmacology , Humans , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, CD/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/chemistry , Animals , Peptide Library , Camelus/immunology , Camelus/genetics , Cell Line, Tumor , Escherichia coli/genetics , T-Lymphocytes/immunology , Gene Expression
2.
Protein Expr Purif ; 221: 106501, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38782081

ABSTRACT

Nanobodies are single-variable domain antibodies with excellent properties, which are evolving as versatile tools to guide cognate antigens in vitro and in vivo for biological research, diagnosis, and treatment. Given their simple structure, nanobodies are readily produced in multiple systems. However, selecting an appropriate expression system is crucial because different conditions might cause proteins to produce different folds or post-translational modifications (PTMs), and these differences often result in different functions. At present, the strategies of PTMs are rarely reported. The GFP nanobody can specifically target the GFP protein. Here, we engineered a GFP nanobody fused with 6 × His tag and Fc tag, respectively, and expressed in bacteria and mammalian cells. The 6 × His-GFP-nanobody was produced from Escherichia coli at high yields and the pull-down assay indicated that it can precipitate the GFP protein. Meanwhile, the Fc-GFP-nanobody can be expressed in HEK293T cells, and the co-immunoprecipitation experiment can trace and target the GFP-tagged protein in vivo. Furthermore, some different PTMs in antigen-binding regions have been identified after using mass spectrometry (MS) to analyze the GFP nanobodies, which are expressed in prokaryotes and eukaryotes. In this study, a GFP nanobody was designed, and its binding ability was verified by using the eukaryotic and prokaryotic protein expression systems. In addition, this GFP nanobody was transformed into a useful instrument for more in-depth functional investigations of GFP fusion proteins. MS was further used to explore the reason for the difference in binding ability, providing a novel perspective for the study of GFP nanobodies and protein expression purification.


Subject(s)
Escherichia coli , Green Fluorescent Proteins , Protein Processing, Post-Translational , Recombinant Fusion Proteins , Single-Domain Antibodies , Humans , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Single-Domain Antibodies/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification , Single-Domain Antibodies/immunology , HEK293 Cells , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/biosynthesis , Protein Engineering/methods , Gene Expression
3.
J Biol Chem ; 299(9): 104927, 2023 09.
Article in English | MEDLINE | ID: mdl-37330175

ABSTRACT

Methicillin-resistant Staphylococcus aureus, or MRSA, is one of the major causative agents of hospital-acquired infections worldwide. Novel antimicrobial strategies efficient against antibiotic-resistant strains are necessary and not only against S. aureus. Among those, strategies that aim at blocking or dismantling proteins involved in the acquisition of essential nutrients, helping the bacteria to colonize the host, are intensively studied. A major route for S. aureus to acquire iron from the host organism is the Isd (iron surface determinant) system. In particular, the hemoglobin receptors IsdH and IsdB located on the surface of the bacterium are necessary to acquire the heme moiety containing iron, making them a plausible antibacterial target. Herein, we obtained an antibody of camelid origin that blocked heme acquisition. We determined that the antibody recognized the heme-binding pocket of both IsdH and IsdB with nanomolar order affinity through its second and third complementary-determining regions. The mechanism explaining the inhibition of acquisition of heme in vitro could be described as a competitive process in which the complementary-determining region 3 from the antibody blocked the acquisition of heme by the bacterial receptor. Moreover, this antibody markedly reduced the growth of three different pathogenic strains of MRSA. Collectively, our results highlight a mechanism for inhibiting nutrient uptake as an antibacterial strategy against MRSA.


Subject(s)
Antibodies, Bacterial , Methicillin-Resistant Staphylococcus aureus , Receptors, Cell Surface , Single-Domain Antibodies , Humans , Anti-Bacterial Agents/pharmacology , Heme/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/therapeutic use , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Single-Domain Antibodies/pharmacology , Staphylococcal Infections/drug therapy , Antigens, Bacterial/immunology , Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Camelids, New World , Animals , Protein Binding/drug effects , Models, Molecular , Molecular Dynamics Simulation
4.
Acta Pharmacol Sin ; 44(5): 1095-1104, 2023 May.
Article in English | MEDLINE | ID: mdl-36418428

ABSTRACT

CKLF (chemokine-like factor)-MARVEL transmembrane domain containing protein 6 (CMTM6) is a novel regulator to maintain the stability of PD-L1. CMTM6 can colocalize and interact with PD-L1 on the recycling endosomes and cell membrane, preventing PD-L1 from lysosome-mediated degradation and proteasome-mediated degradation thus increasing the half-life of PD-L1 on the cell membrane. The difficulties in obtaining stable full-length PD-L1 and CMTM6 proteins hinder the research on their structures, function as well as related drug development. Using lauryl maltose neopentyl glycol (LMNG) as the optimized detergent and a cell membrane mimetic strategy, we assembled a stable membrane-bound full-length CMTM6-PD-L1 complex with amphipol A8-35. When the PD-1/PD-L1-CMTM6 interactions were analyzed, we found that CMTM6 greatly enhanced the binding and delayed the dissociation of PD-1/PD-L1, thus affecting immunosuppressive signaling and anti-apoptotic signaling. We then used the CMTM6-PD-L1 complex as immunogens to generate immune repertoires in camels, and identified a functional anti-CMTM6 nanobody, called 1A5. We demonstrated that the anti-CMTM6 nanobody greatly decreased T-cell immunosuppression and promoted apoptotic susceptibility of tumor cells in vitro, and mainly relied on the cytotoxic effect of CD8+ T-cells to exert tumor growth inhibitory effects in CT26 tumor-bearing mice. In conclusion, the stable membrane-bound full-length CMTM6-PD-L1 complex has been successfully used in studying PD-1/PD-L1-CMTM6 interactions and CMTM6-targeting drug development, suggesting CMTM6 as a novel tumor immunotherapy target.


Subject(s)
B7-H1 Antigen , MARVEL Domain-Containing Proteins , Neoplasms , Single-Domain Antibodies , Animals , Mice , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/metabolism , MARVEL Domain-Containing Proteins/immunology , MARVEL Domain-Containing Proteins/metabolism , Protein Engineering/methods , Single-Domain Antibodies/biosynthesis
5.
Signal Transduct Target Ther ; 7(1): 44, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140196

ABSTRACT

The wide transmission and host adaptation of SARS-CoV-2 have led to the rapid accumulation of mutations, posing significant challenges to the effectiveness of vaccines and therapeutic antibodies. Although several neutralizing antibodies were authorized for emergency clinical use, convalescent patients derived natural antibodies are vulnerable to SARS-CoV-2 Spike mutation. Here, we describe the screen of a panel of SARS-CoV-2 receptor-binding domain (RBD) targeted nanobodies (Nbs) from a synthetic library and the design of a biparatopic Nb, named Nb1-Nb2, with tight affinity and super-wide neutralization breadth against multiple SARS-CoV-2 variants of concern. Deep-mutational scanning experiments identify the potential binding epitopes of the Nbs on the RBD and demonstrate that biparatopic Nb1-Nb2 has a strong escape-resistant feature against more than 60 tested RBD amino acid substitutions. Using pseudovirion-based and trans-complementation SARS-CoV-2 tools, we determine that the Nb1-Nb2 broadly neutralizes multiple SARS-CoV-2 variants at sub-nanomolar levels, including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Lambda (C.37), Kappa (B.1.617.1), and Mu (B.1.621). Furthermore, a heavy-chain antibody is constructed by fusing the human IgG1 Fc to Nb1-Nb2 (designated as Nb1-Nb2-Fc) to improve its neutralization potency, yield, stability, and potential half-life extension. For the new Omicron variant (B.1.1.529) that harbors unprecedented multiple RBD mutations, Nb1-Nb2-Fc keeps a firm affinity (KD < 1.0 × 10-12 M) and strong neutralizing activity (IC50 = 1.46 nM for authentic Omicron virus). Together, we developed a tetravalent biparatopic human heavy-chain antibody with ultrapotent and broad-spectrum SARS-CoV-2 neutralization activity which highlights the potential clinical applications.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Recombinant Fusion Proteins/pharmacology , SARS-CoV-2/drug effects , Single-Domain Antibodies/pharmacology , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/genetics , Antibodies, Viral/biosynthesis , Antibodies, Viral/genetics , Antibody Affinity , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Epitopes/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin Fc Fragments/genetics , Models, Molecular , Neutralization Tests , Protein Binding/drug effects , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/genetics , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
6.
FASEB J ; 36(4): e22222, 2022 04.
Article in English | MEDLINE | ID: mdl-35218573

ABSTRACT

Cellular uptake of vitamin B12 in humans is mediated by the endocytosis of the B12 carrier protein transcobalamin (TC) via its cognate cell surface receptor TCblR, encoded by the CD320 gene. Because CD320 expression is associated with the cell cycle and upregulated in highly proliferating cells including cancer cells, this uptake route is a potential target for cancer therapy. We developed and characterized four camelid nanobodies that bind holo-TC (TC in complex with B12 ) or the interface of the human holo-TC:TCblR complex with nanomolar affinities. We determined X-ray crystal structures of these nanobodies bound to holo-TC:TCblR, which enabled us to map their binding epitopes. When conjugated to the model toxin saporin, three of our nanobodies caused growth inhibition of HEK293T cells and therefore have the potential to inhibit the growth of human cancer cells. We visualized the cellular binding and endocytic uptake of the most potent nanobody (TC-Nb4) using fluorescent light microscopy. The co-crystal structure of holo-TC:TCblR with another nanobody (TC-Nb34) revealed novel features of the interface of TC and the LDLR-A1 domain of TCblR, rationalizing the decrease in the affinity of TC-B12 binding caused by the Δ88 mutation in CD320.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoconjugates/pharmacology , Receptors, Cell Surface/metabolism , Saporins/chemistry , Single-Domain Antibodies/chemistry , Transcobalamins/metabolism , Vitamin B 12/metabolism , Animals , Antibodies, Monoclonal/immunology , Camelids, New World , Cell Cycle , Cell Proliferation , HEK293 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunotoxins/chemistry , Immunotoxins/pharmacology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Saporins/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/immunology
7.
Int J Biol Macromol ; 193(Pt B): 1130-1137, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34699899

ABSTRACT

Monoclonal antibodies have contributed to improving the treatment of several diseases. However, limitations related to pharmacokinetic parameters and production costs have instigated the search for alternative products. Camelids produce functional immunoglobulins G devoid of light chains and CH1 domains, in which the antigenic recognition site is formed by a single domain called VHH or nanobody. VHHs' small size and similarity to the human VH domain contribute to high tissue penetration and low immunogenicity. In addition, VHHs provide superior antigen recognition compared to human antibodies, better solubility and stability. Due to these characteristics and the possibility of obtaining gene-encoding VHHs, applications of this biological tool, whether as a monomer or in related recombinant constructs, have been reported. To ensure antibody efficacy and cost-effectiveness, strategies for their expression, either using prokaryotic or eukaryotic systems, have been utilized. Plant-based expression systems are useful for VHH related constructs that require post-translational modifications. This system has exhibited versatility, low-cost upstream production, and safety. This article presents the main advances associated to the heterologous expression of VHHs in plant systems. Besides, we show insights related to the use of VHHs as a strategy for plant pathogen control and a tool for genomic manipulation in plant systems.


Subject(s)
Gene Expression , Plants, Genetically Modified , Plants , Single-Domain Antibodies , Animals , Humans , Plants/genetics , Plants/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/genetics
8.
Biochem Biophys Res Commun ; 581: 38-45, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34653677

ABSTRACT

A detrimental role of the receptor for the advanced glycation end product (RAGE) has been identified in the immune response, and various pathological conditions and its V and C1 domains in the extracellular region of RAGE are believed to be the main ligand-binding domains. Consequently, specific inhibitors targeting those domains could be of clinical value in fighting against the pathological condition associated with RAGE over-activation. Single-domain antibodies, also called nanobodies (Nbs), are antibody fragments engineered from the heavy-chain only antibodies found in camelids, which offer a range of advantages in therapy. In this study, we report the development and characterization of the V-C1 domain-specific Nbs. Three Nbs (3CNB, 4BNB, and 5ENB) targeting V-C1 domain of human RAGE were isolated from an immunized alpaca using a phage display. All of these Nbs revealed high thermostability. 3CNB, 4BNB, and 5ENB bind to V-C1 domain with a dissociation constant (KD) of 27.25, 39.37, and 47.85 nM, respectively, using Isothermal Titration Calorimetry (ITC). After homodimerization using human IgG1-Fc fusion, their binding affinity improved to 0.55, 0.62, and 0.41 nM, respectively, using Surface Plasmon Resonance (SPR). Flow cytometry showed all the Fc fusions Nbs can bind to human RAGE expressed on the cell surface. Competitive ELISA further confirmed their V-C1-hS100B blocking ability in solution, providing insights into the applicability of Nbs in treating RAGE-associated diseases.


Subject(s)
Glycation End Products, Advanced/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Receptor for Advanced Glycation End Products/chemistry , Recombinant Fusion Proteins/chemistry , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Binding Sites , Camelids, New World , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/immunology , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Peptide Library , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , Protein Multimerization , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/isolation & purification
9.
Biochim Biophys Acta Gen Subj ; 1865(11): 129974, 2021 11.
Article in English | MEDLINE | ID: mdl-34343644

ABSTRACT

Background Since December 2019, the newly emerged SARS-CoV-2 virus continues to infect humans and many people died from severe Covid-19 during the last 2 years worldwide. Different approaches are being used for treatment of this infection and its consequences, but limited results have been achieved and new therapeutics are still needed. One of the most interesting biotherapeutics in this era are Nanobodies which have shown very promising results in recent researches. Scope of review Here, we have reviewed the potentials of Nanobodies in Covid-19 treatment. We have also discussed the properties of these biotherapeutics that make them very suitable for pulmonary drug delivery, which seems to be very important route of administration in this disease. Major conclusion Nanobodies with their special biological and biophysical characteristics and their resistance against harsh manufacturing condition, can be considered as promising, targeted biotherapeutics which can be administered by pulmonary delivery pharmaceutical systems against Covid-19. General significance Covid-19 has become a global problem during the last two years and with emerging mutant strains, prophylactic and therapeutic approaches are still highly needed. Nanobodies with their specific properties can be considered as valuable and promising candidates in Covid-19 therapy.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antiviral Agents/therapeutic use , COVID-19/therapy , Immunologic Factors/therapeutic use , SARS-CoV-2/drug effects , Single-Domain Antibodies/therapeutic use , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Neutralizing/isolation & purification , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , COVID-19/immunology , COVID-19/virology , Camelus , Drug Delivery Systems , Humans , Immune Sera/chemistry , Immunologic Factors/biosynthesis , Immunologic Factors/isolation & purification , Lung/drug effects , Lung/immunology , Lung/virology , Molecular Targeted Therapy/methods , Peptide Library , Protein Binding/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
10.
ACS Synth Biol ; 10(8): 2049-2059, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34337948

ABSTRACT

Gram-negative bacteria are endowed with complex outer membrane (OM) structures that allow them to both interact with other organisms and attach to different physical structures. However, the design of reliable bacterial coatings of solid surfaces is still a considerable challenge. In this work, we report that ectopic expression of a fibrinogen-specific nanobody on the envelope of Pseudomonas putida cells enables controllable formation of a bacterial monolayer strongly bound to an antigen-coated support. To this end, either the wild type or a surface-naked derivative of P. putida was engineered to express a hybrid between the ß-barrel of an intimin-type autotransporter inserted in the outer membrane and a nanobody (VHH) moiety that targets fibrinogen as its cognate interaction partner. The functionality of the thereby presented VHH and the strength of the resulting cell attachment to a solid surface covered with the cognate antigen were tested and parametrized with Quartz Crystal Microbalance technology. The results not only demonstrated the value of using bacteria with reduced OM complexity for efficient display of artificial adhesins, but also the potential of this approach to engineer specific bacterial coverings of predetermined target surfaces.


Subject(s)
Cell Surface Display Techniques , Pseudomonas putida , Recombinant Fusion Proteins , Single-Domain Antibodies , Pseudomonas putida/genetics , Pseudomonas putida/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/genetics
11.
Protein Expr Purif ; 185: 105906, 2021 09.
Article in English | MEDLINE | ID: mdl-33991675

ABSTRACT

Nanobodies are single-domain antibody constructs derived from the variable regions of heavy chain only (VHH) camelid IgGs. Their small size and single gene format make them amenable to various molecular biology applications that require a protein affinity-based approach. These features, in addition to their high solubility, allows their periplasmic expression, extraction and purification in E. coli systems with relative ease, using standardized protocols. However, some Nanobodies are recalcitrant to periplasmic expression, extraction and purification within E. coli systems. To improve their expression would require either a change in the expression host, vector or an increased scale of expression, all of which entail an increase in the complexity of their expression, and production cost. However, as shown here, specific changes in the existing standard E. coli culture protocol, aimed at reducing breakdown of selective antibiotic pressure, increasing the initial culture inoculum and improving transport to the periplasmic space, rescued the expression of several such refractory Nanobodies. The periplasmic extraction protocol was also changed to ensure efficient osmolysis, prevent both protein degradation and prevent downstream chelation of Ni2+ ions during IMAC purification. Adoption of this protocol will lead to an improvement of the expression of Nanobodies in general, and specifically, those that are recalcitrant.


Subject(s)
Escherichia coli/metabolism , Periplasm/metabolism , Recombinant Proteins/isolation & purification , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Cloning, Molecular , Culture Media/chemistry , Culture Media/pharmacology , Escherichia coli/genetics , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Osmotic Pressure , Periplasm/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Single-Domain Antibodies/genetics , Single-Domain Antibodies/isolation & purification
12.
Clin Biochem ; 93: 80-89, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33831386

ABSTRACT

OBJECTIVES: Mutations in the gene encoding the glycogen phosphatase laforin result in the fatal childhood dementia Lafora disease (LD). A cellular hallmark of LD is cytoplasmic, hyper-phosphorylated, glycogen-like aggregates called Lafora bodies (LBs) that form in nearly all tissues and drive disease progression. Additional tools are needed to define the cellular function of laforin, understand the pathological role of laforin in LD, and determine the role of glycogen phosphate in glycogen metabolism. In this work, we present the generation and characterization of laforin nanobodies, with one being a laforin inhibitor. DESIGN AND METHODS: We identify multiple classes of specific laforin-binding nanobodies and determine their binding epitopes using hydrogen deuterium exchange (HDX) mass spectrometry. Using para-nitrophenyl phosphate (pNPP) and a malachite gold-based assay specific for glucan phosphatase activity, we assess the inhibitory effect of one nanobody on laforin's catalytic activity. RESULTS: Six families of laforin nanobodies are characterized and their epitopes mapped. One nanobody is identified and characterized that serves as an inhibitor of laforin's phosphatase activity. CONCLUSIONS: The six generated and characterized laforin nanobodies, with one being a laforin inhibitor, are an important set of tools that open new avenues to define unresolved glycogen metabolism questions.


Subject(s)
Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/chemistry , Animals , Biological Assay , Camelids, New World , Chromatography, Gel , Enzyme Inhibitors/pharmacology , Epitope Mapping , Glycogen/metabolism , Gold/chemistry , Humans , Hydrogen Deuterium Exchange-Mass Spectrometry , Lafora Disease/enzymology , Models, Molecular , Nitrophenols/chemistry , Organometallic Compounds/chemistry , Organophosphorus Compounds/chemistry , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Single-Domain Antibodies/isolation & purification
13.
Protein Expr Purif ; 184: 105888, 2021 08.
Article in English | MEDLINE | ID: mdl-33857600

ABSTRACT

Anti-EGFR nanobodies have been successfully applied as antitumor moieties in the photodynamic therapy and drug delivery systems. But the yields of nanobodies were still limited due to the volumetric capacity of the periplasmic compartments and inclusion bodies of Escherichia coli. A comparative study of Pichia pastoris and Escherichia coli was done through characterizing their products. Nanobody 7D12 and 7D12-9G8 were successfully expressed in Pichia pastoris with 6-13.6-fold higher yield. Both two types of nanobodies had internalization ability to be developed as antitumor moieties.


Subject(s)
Antineoplastic Agents, Immunological , Escherichia coli , Neoplasm Proteins , Saccharomycetales , Single-Domain Antibodies , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Saccharomycetales/genetics , Saccharomycetales/metabolism , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/immunology , Single-Domain Antibodies/isolation & purification , Single-Domain Antibodies/pharmacology
14.
Biochem Biophys Res Commun ; 549: 105-112, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33667707

ABSTRACT

Escherichia coli is one of the most popularly used hosts to produce recombinant proteins. Most recombinant proteins are produced in the cytoplasm and periplasm, requiring multiple steps to extract and purify recombinant proteins. The Serratia marcescens Lip system (LipB-LipC-LipD) is a type 1 secretion system that selectively secretes LipA from the intracellular to extracellular space in a single step. This study aimed to establish a secretory production system for nanobodies, camelid-derived small molecule antibody fragments, using the S. marcescens Lip system. Surprisingly, E. coli harboring only LipC, a membrane fusion protein of the Lip system, could secrete an anti-green fluorescent protein (GFP)-Nb, a nanobody against GFP, without the addition of a long amino acid sequence. The LipC-based secretion system recognized the Val-Thr-Val sequence at the C-terminus of the nanobody. Finally, Strep-tagged anti-GFP-Nb was purified from culture supernatants of E. coli harboring LipC by Strep-affinity chromatography at a final yield of >5 mg per liter of culture supernatant. These results potently supported that the S. marcescens LipC-based secretion system has the potential to establish an efficient secretory production system for nanobodies.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/metabolism , Serratia marcescens/metabolism , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Antigens/metabolism , Camelus , Culture Media , Green Fluorescent Proteins/metabolism , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/isolation & purification
15.
Sci Rep ; 11(1): 3126, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542302

ABSTRACT

Influenza H7N9 virus continues to cause infections in humans and represents a significant pandemic risk. During the most recent 5th epidemic wave in 2016/17 two distinct lineages with increased human infections and wider geographical spread emerged. In preparation for any future adaptations, broadly reactive antibodies against H7N9 are required for surveillance, therapy and prophylaxis. In this study we have isolated a panel of nanobodies (Nbs) with broad reactivity across H7 influenza strains, including H7N9 strains between 2013 and 2017. We also describe Nbs capable of distinguishing between the most recent high and low pathogenicity Yangtze River Delta lineage H7N9 strains. Nanobodies were classified into 5 distinct groups based on their epitope footprint determined using yeast display and mutational scanning. The epitope footprint of Nbs capable of distinguishing high pathogenic (HP) A/Guangdong/17SF003/2016 from low pathogenic (LP) A/Hong Kong/125/2017 (H7N9) were correlated to natural sequence divergence in the head domain at lysine 164. Several Nbs binding to the head domain were capable of viral neutralisation. The potency of one nanobody NB7-14 could be increased over 1000-fold to 113 pM by linking two Nbs together. Nbs specific for distinct epitopes on H7N9 may be useful for surveillance or therapy in human or veterinary settings.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N9 Subtype/drug effects , Peptide Library , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Binding Sites , Birds/virology , Epitopes/chemistry , Epitopes/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza in Birds/immunology , Influenza in Birds/prevention & control , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/transmission , Influenza, Human/virology , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Single-Domain Antibodies/isolation & purification
16.
Biochem J ; 478(3): 579-595, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33480416

ABSTRACT

Surface-associated proteins play critical roles in the Plasmodium parasite life cycle and are major targets for vaccine development. The 6-cysteine (6-cys) protein family is expressed in a stage-specific manner throughout Plasmodium falciparum life cycle and characterized by the presence of 6-cys domains, which are ß-sandwich domains with conserved sets of disulfide bonds. Although several 6-cys family members have been implicated to play a role in sexual stages, mosquito transmission, evasion of the host immune response and host cell invasion, the precise function of many family members is still unknown and structural information is only available for four 6-cys proteins. Here, we present to the best of our knowledge, the first crystal structure of the 6-cys protein Pf12p determined at 2.8 Šresolution. The monomeric molecule folds into two domains, D1 and D2, both of which adopt the canonical 6-cys domain fold. Although the structural fold is similar to that of Pf12, its paralog in P. falciparum, we show that Pf12p does not complex with Pf41, which is a known interaction partner of Pf12. We generated 10 distinct Pf12p-specific nanobodies which map into two separate epitope groups; one group which binds within the D2 domain, while several members of the second group bind at the interface of the D1 and D2 domain of Pf12p. Characterization of the structural features of the 6-cys family and their associated nanobodies provide a framework for generating new tools to study the diverse functions of the 6-cys protein family in the Plasmodium life cycle.


Subject(s)
Antigens, Protozoan/chemistry , Single-Domain Antibodies/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Binding Sites , Blotting, Western , Camelids, New World/immunology , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Interferometry , Models, Molecular , Peptide Fragments/genetics , Peptide Fragments/metabolism , Plasmodium falciparum/metabolism , Protein Conformation , Protein Domains , Protein Interaction Mapping , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification
17.
Methods Mol Biol ; 2248: 91-107, 2021.
Article in English | MEDLINE | ID: mdl-33185870

ABSTRACT

Systemic cytokine inhibition may be an effective therapeutic strategy for several autoimmune diseases. However, recent studies suggest that tissue or cell type-specific targeting of certain cytokines, including TNF, may have distinct advantages and show fewer side effects. Here we describe protocols for generating and testing bispecific cytokine inhibitors using variable domain of single-chain antibodies from Camelidae (VHH) with a focus on cell-specific TNF inhibitors.


Subject(s)
Immunoglobulin Heavy Chains , Immunoglobulin Variable Region , Single-Domain Antibodies/biosynthesis , Tumor Necrosis Factor Inhibitors , Antibody Affinity , Antibody Specificity , Cytokines/biosynthesis , Flow Cytometry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/isolation & purification , Single-Domain Antibodies/pharmacology , Tumor Necrosis Factor Inhibitors/chemistry , Tumor Necrosis Factor Inhibitors/isolation & purification , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factors/chemistry , Tumor Necrosis Factors/metabolism
18.
Biomolecules ; 10(12)2020 12 09.
Article in English | MEDLINE | ID: mdl-33317184

ABSTRACT

The Zika virus was introduced in Brazil in 2015 and, shortly after, spread all over the Americas. Nowadays, it remains present in more than 80 countries and represents a major threat due to some singularities among other flaviviruses. Due to its easy transmission, high percentage of silent cases, the severity of its associated complications, and the lack of prophylactic methods and effective treatments, it is essential to develop reliable and rapid diagnostic tests for early containment of the infection. Nonstructural protein 1 (NS1), a glycoprotein involved in all flavivirus infections, is secreted since the beginning of the infection into the blood stream and has proven to be a valuable biomarker for the early diagnosis of other flaviviral infections. Here, we describe the development of a highly sensitive nanobody ELISA for the detection of the NS1 protein in serum samples. Nanobodies were selected from a library generated from a llama immunized with Zika NS1 (ZVNS1) by a two-step high-throughput screening geared to identify the most sensitive and specific nanobody pairs. The assay was performed with a sub-ng/mL detection limit in the sera and showed excellent reproducibility and accuracy when validated with serum samples spiked with 0.80, 1.60, or 3.10 ng/mL of ZVNS1. Furthermore, the specificity of the developed ELISA was demonstrated using a panel of flavivirus' NS1 proteins; this is of extreme relevance in countries endemic for more than one flavivirus. Considering that the nanobody sequences are provided, the assay can be reproduced in any laboratory at low cost, which may help to strengthen the diagnostic capacity of the disease even in low-resource countries.


Subject(s)
Antibodies, Viral/chemistry , Enzyme-Linked Immunosorbent Assay/standards , Single-Domain Antibodies/chemistry , Viral Nonstructural Proteins/blood , Zika Virus Infection/diagnosis , Zika Virus/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/isolation & purification , Camelids, New World , Enzyme-Linked Immunosorbent Assay/methods , Escherichia coli/genetics , Humans , Limit of Detection , Peptide Library , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/isolation & purification , Uruguay , Zika Virus Infection/blood , Zika Virus Infection/immunology , Zika Virus Infection/virology
19.
Biotechnol Bioeng ; 117(12): 3835-3848, 2020 12.
Article in English | MEDLINE | ID: mdl-32808670

ABSTRACT

Growth decoupling can be used to optimize the production of biochemicals and proteins in cell factories. Inhibition of excess biomass formation allows for carbon to be utilized efficiently for product formation instead of growth, resulting in increased product yields and titers. Here, we used CRISPR interference to increase the production of a single-domain antibody (sdAb) by inhibiting growth during production. First, we screened 21 sgRNA targets in the purine and pyrimidine biosynthesis pathways and found that the repression of 11 pathway genes led to the increased green fluorescent protein production and decreased growth. The sgRNA targets pyrF, pyrG, and cmk were selected and further used to improve the production of two versions of an expression-optimized sdAb. Proteomics analysis of the sdAb-producing pyrF, pyrG, and cmk growth decoupling strains showed significantly decreased RpoS levels and an increase of ribosome-associated proteins, indicating that the growth decoupling strains do not enter stationary phase and maintain their capacity for protein synthesis upon growth inhibition. Finally, sdAb production was scaled up to shake-flask fermentation where the product yield was improved 2.6-fold compared to the control strain with no sgRNA target sequence. An sdAb content of 14.6% was reached in the best-performing pyrG growth decoupling strain.


Subject(s)
CRISPR-Cas Systems , Escherichia coli , Metabolic Engineering , Nucleotides , Single-Domain Antibodies/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Nucleotides/biosynthesis , Nucleotides/genetics , Single-Domain Antibodies/genetics
20.
J Immunol Methods ; 483: 112811, 2020 08.
Article in English | MEDLINE | ID: mdl-32569598

ABSTRACT

Due to the technical innovations in generating bispecific antibodies (BsAbs) in recent years, BsAbs have become important reagents for diagnostic and therapeutic applications. However, the difficulty of producing a heterodimer consisting of two different arms with high yield and purity constituted a major limitation for their application in academic and clinical settings. Here, we describe a novel Fc-containing BsAb format (Fab × sdAb-Fc) composed of a conventional antigen-binding fragment (Fab), and a single domain antibody (sdAb), which avoids heavy-light chain mis-pairing during antibody assembly. In this study, the Fab x sdAb-Fc BsAbs were efficiently produced by three widely used heavy-heavy chain heterodimerization methods: Knobs-into-holes (KIH), Charge-pairs (CP) and controlled Fab-arm exchange (cFAE), respectively. The novel Fab x sdAb-Fc format provided a rapid and efficient strategy to generate BsAb with high purity and a unique possibility to further purify desired BsAbs from undesired antibodies based on molecular weight (MW). Compared to conventional BsAb formats, the advantages of Fab x sdAb-Fc format may thus provide a straightforward opportunity to apply bispecific antibody principles to research and development of novel targets and pathways in diseases such as cancer and autoimmunity.


Subject(s)
Antibodies, Bispecific/immunology , ErbB Receptors/immunology , Glutamate Carboxypeptidase II/immunology , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Membrane Glycoproteins/immunology , Single-Domain Antibodies/immunology , Animals , Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/genetics , Antibody Specificity , CHO Cells , Cricetulus , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin Fc Fragments/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Weight , Mutation , Proof of Concept Study , Protein Multimerization , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/genetics
SELECTION OF CITATIONS
SEARCH DETAIL