Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37.805
Filter
1.
Front Immunol ; 15: 1407567, 2024.
Article in English | MEDLINE | ID: mdl-39100677

ABSTRACT

Introduction: NK cells can mediate tumor cell killing by natural cytotoxicity and by antibody-dependent cell-mediated cytotoxicity (ADCC), an anti-tumor mechanism mediated through the IgG Fc receptor CD16A (FcγRIIIA). CD16A polymorphisms conferring increased affinity for IgG positively correlate with clinical outcomes during monoclonal antibody therapy for lymphoma, linking increased binding affinity with increased therapeutic potential via ADCC. We have previously reported on the FcγR fusion CD64/16A consisting of the extracellular region of CD64 (FcγRI), a high-affinity Fc receptor normally expressed by myeloid cells, and the transmembrane/cytoplasmic regions of CD16A, to create a highly potent and novel activating fusion receptor. Here, we evaluate the therapeutic potential of engineered induced pluripotent stem cell (iPSC)-derived NK (iNK) cells expressing CD64/16A as an "off-the-shelf", antibody-armed cellular therapy product with multi-antigen targeting potential. Methods: iNK cells were generated from iPSCs engineered to express CD64/16A and an interleukin (IL)-15/IL-15Rα fusion (IL-15RF) protein for cytokine independence. iNK cells and peripheral blood NK cells were expanded using irradiated K562-mbIL21-41BBL feeder cells to examine in in vitro and in vivo assays using the Raji lymphoma cell line. ADCC was evaluated in real-time by IncuCyte assays and using a xenograft mouse model with high circulating levels of human IgG. Results: Our data show that CD64/16A expressing iNK cells can mediate potent anti-tumor activity against human B cell lymphoma. In particular, (i) under suboptimal conditions, including low antibody concentrations and low effector-to-target ratios, iNK-CD64/16A cells mediate ADCC, (ii) iNK-CD64/16A cells can be pre-loaded with tumor-targeting antibodies (arming) to elicit ADCC, (iii) armed iNK-CD64/16A cells can be repurposed with additional antibodies to target new tumor antigens, and (iv) cryopreserved, armed iNK-CD64/16A are capable of sustained ADCC in a tumor xenograft model under saturating levels of human IgG. Discussion: iNK-CD64/16A cells allow for a flexible use of antibodies (antibody arming and antibody targeting), and an "off-the-shelf" platform for multi-antigen recognition to overcome limitations of adoptive cell therapies expressing fixed antigen receptors leading to cancer relapse due to antigen escape variants.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Antigens, Neoplasm , Induced Pluripotent Stem Cells , Killer Cells, Natural , Lymphoma , Receptors, IgG , Xenograft Model Antitumor Assays , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, IgG/genetics , Humans , Animals , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Lymphoma/therapy , Lymphoma/immunology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/immunology , Antigens, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line, Tumor , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Mice, SCID
2.
Nat Commun ; 15(1): 6512, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095395

ABSTRACT

Many disease resistance genes have been introgressed into wheat from its wild relatives. However, reduced recombination within the introgressed segments hinders the cloning of the introgressed genes. Here, we have cloned the powdery mildew resistance gene Pm13, which is introgressed into wheat from Aegilops longissima, using a method that combines physical mapping with radiation-induced chromosomal aberrations and transcriptome sequencing analysis of ethyl methanesulfonate (EMS)-induced loss-of-function mutants. Pm13 encodes a kinase fusion protein, designated MLKL-K, with an N-terminal domain of mixed lineage kinase domain-like protein (MLKL_NTD domain) and a C-terminal serine/threonine kinase domain bridged by a brace. The resistance function of Pm13 is validated through transient and stable transgenic complementation assays. Transient over-expression analyses in Nicotiana benthamiana leaves and wheat protoplasts reveal that the fragment Brace-Kinase122-476 of MLKL-K is capable of inducing cell death, which is dependent on a functional kinase domain and the three α-helices in the brace region close to the N-terminus of the kinase domain.


Subject(s)
Aegilops , Ascomycota , Disease Resistance , Plant Diseases , Plant Proteins , Triticum , Triticum/microbiology , Triticum/genetics , Plant Diseases/microbiology , Plant Diseases/immunology , Plant Diseases/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Disease Resistance/genetics , Aegilops/genetics , Aegilops/metabolism , Plants, Genetically Modified , Protein Kinases/metabolism , Protein Kinases/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Nicotiana/genetics , Nicotiana/microbiology , Plant Leaves/microbiology , Plant Leaves/genetics , Plant Leaves/metabolism , Gene Expression Regulation, Plant
4.
Viruses ; 16(7)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39066255

ABSTRACT

A wide range of virus-like particles (VLPs) is extensively employed as carriers to display various antigens for vaccine development to fight against different infections. The plant-produced truncated variant of the hepatitis E virus (HEV) coat protein is capable of forming VLPs. In this study, we demonstrated that recombinant fusion proteins comprising truncated HEV coat protein with green fluorescent protein (GFP) or four tandem copies of the extracellular domain of matrix protein 2 (M2e) of influenza A virus inserted at the Tyr485 position could be efficiently expressed in Nicotiana benthamiana plants using self-replicating vector based on the potato virus X genome. The plant-produced fusion proteins in vivo formed VLPs displaying GFP and 4M2e. Therefore, HEV coat protein can be used as a VLP carrier platform for the presentation of relatively large antigens comprising dozens to hundreds of amino acids. Furthermore, plant-produced HEV particles could be useful research tools for the development of recombinant vaccines against influenza.


Subject(s)
Antigen Presentation , Capsid Proteins , Hepatitis E virus , Nicotiana , Recombinant Fusion Proteins , Viral Matrix Proteins , Hepatitis E virus/immunology , Hepatitis E virus/genetics , Nicotiana/virology , Nicotiana/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Plants, Genetically Modified , Influenza A virus/immunology , Influenza A virus/genetics , Hepatitis E/immunology , Hepatitis E/prevention & control , Hepatitis E/virology , Viroporin Proteins
5.
Protein Sci ; 33(8): e5066, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39074259

ABSTRACT

Affinity precipitation is an attractive method for protein purification due to its many advantages, including the rapid capture of target proteins, simple processing, high specificity, and ease of scale-up. We previously reported a robust antibody purification method using Ca2+-dependent precipitation of ZZ-hCSQ2, a fusion protein of human calsequestrin 2, and the antibody-binding protein ZZ. However, the stability of this fusion protein was not sufficiently high for industrial use because the antibody recovery yield decreased to 60% after being reused 10 times. To identify a more stable calsequestrin (CSQ), we calculated Rosetta energy values for the folding stabilities of various CSQ homologs and selected human CSQ1 (hCSQ1) with lowest energy value (-992.6) as the new CSQ platform. We also identified that the linker sequence between ZZ and CSQ was vulnerable to proteases and alkaline pH by N-terminal protein sequencing. Therefore, we changed the linker to four asparagine (4N) sequences, which were shorter and less flexible than the previous glycine-rich linker. The new version of ZZ-CSQ, ZZ-4N-hCSQ1, was stable in a protease-containing conditioned medium obtained from the cultured Chinese hamster ovary cell or high pH condition (0.1M sodium hydroxide) for more than 5 days and could be reused at least 25 times for antibody purification without loss of recovery yield. The antibodies purified by ZZ-4N-hCSQ1 precipitation also showed greater purity (~33.6-fold lower host cell DNA and ~6.4-fold lower host cell protein) than those purified by protein A chromatography. These data suggest that ZZ-4N-hCSQ1 precipitation is more efficient and can achieve cost-effectiveness of up to 12.5-fold cheaper than previous antibody purification methods and can lower the production costs of therapeutic antibodies.


Subject(s)
Calcium , Humans , Calcium/chemistry , Calsequestrin/chemistry , Calsequestrin/genetics , Calsequestrin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Protein Stability , Animals , CHO Cells , Cricetulus , Chemical Precipitation
6.
PLoS One ; 19(7): e0307713, 2024.
Article in English | MEDLINE | ID: mdl-39058754

ABSTRACT

A DNA cassette assembly method is described which utilizes inexpensive oligomers no longer than 40 nt in length. The five-segment cassettes have 20 nt overlaps which give an effective length of 80 nt, making it possible to code for peptides up to 20 amino acids long. The cassettes have three phosphate free nicks, which can be successfully inserted into plasmid DNA and used to transform E. coli. The nicks are repaired in vivo by an unknown mechanism. Insertions are not successful for cassettes with greater than three nicks. A procedure is provided for rapid turnaround from DNA design to peptides, which are easily isolated as C-terminal fusions with GFP. The technique generally gives the expected sequence, with errors which occur about 1% of the time. Several representative DNA inserts are described which illustrate the method, as well as chemical details on the new peptides coded for. The peptides can be readily mutated to make it possible to understand how polar and aromatic residues affect GFP-fusion solubility, and how histidine residues can be strategically placed in a peptide for good IMAC retention. The method can be used to explore a large number of new designed peptides as fusion products quickly and economically.


Subject(s)
DNA , Escherichia coli , Peptides , Peptides/chemistry , Peptides/genetics , DNA/genetics , DNA/chemistry , Escherichia coli/genetics , Plasmids/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/chemistry , Mutagenesis, Insertional , Amino Acid Sequence , Base Sequence
7.
Biosci Rep ; 44(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39011584

ABSTRACT

Botulinum neurotoxins (BoNTs) are valuable tools to unveil molecular mechanisms of exocytosis in neuronal and non-neuronal cells due to their peptidase activity on exocytic isoforms of SNARE proteins. They are produced by Clostridia as single-chain polypeptides that are proteolytically cleaved into light, catalytic domains covalently linked via disulfide bonds to heavy, targeting domains. This format of two subunits linked by disulfide bonds is required for the full neurotoxicity of BoNTs. We have generated a recombinant version of BoNT/B that consists of the light chain of the toxin fused to the protein transduction domain of the human immunodeficiency virus-1 (TAT peptide) and a hexahistidine tag. His6-TAT-BoNT/B-LC, expressed in Escherichia coli and purified by affinity chromatography, penetrated membranes and exhibited strong enzymatic activity, as evidenced by cleavage of the SNARE synaptobrevin from rat brain synaptosomes and human sperm cells. Proteolytic attack of synaptobrevin hindered exocytosis triggered by a calcium ionophore in the latter. The novel tool reported herein disrupts the function of a SNARE protein within minutes in cells that may or may not express the receptors for the BoNT/B heavy chain, and without the need for transient transfection or permeabilization.


Subject(s)
Botulinum Toxins, Type A , Exocytosis , Animals , Humans , Rats , Botulinum Toxins, Type A/metabolism , Botulinum Toxins, Type A/genetics , Botulinum Toxins, Type A/isolation & purification , SNARE Proteins/metabolism , SNARE Proteins/genetics , Male , Synaptosomes/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/genetics , Cell Membrane Permeability/drug effects , Botulinum Toxins/metabolism , Botulinum Toxins/genetics , Botulinum Toxins/chemistry , Botulinum Toxins/isolation & purification
8.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062851

ABSTRACT

The requirement for fast and dependable protein purification methods is constant, either for functional studies of natural proteins or for the production of biotechnological protein products. The original procedure has to be formulated for each individual protein, and this demanding task was significantly simplified by the introduction of affinity tags. Helicobacter pylori adenylosuccinate synthetase (AdSS) is present in solution in a dynamic equilibrium of monomers and biologically active homodimers. The addition of the His6-tag on the C-terminus (C-His-AdSS) was proven to have a negligible effect on the characteristics of this enzyme. This paper shows that the same enzyme with the His6-tag fused on its N-terminus (N-His-AdSS) has a high tendency to precipitate. Circular dichroism and X-ray diffraction studies do not detect any structural change that could explain this propensity. However, the dynamic light scattering, differential scanning fluorimetry, and analytical ultracentrifugation measurements indicate that the monomer of this construct is prone to aggregation, which shifts the equilibrium towards the insoluble precipitant. In agreement, enzyme kinetics measurements showed reduced enzyme activity, but preserved affinity for the substrates, in comparison with the wild-type and C-His-AdSS. The presented results reinforce the notion that testing the influence of the tag on protein properties should not be overlooked.


Subject(s)
Adenylosuccinate Synthase , Helicobacter pylori , Histidine , Helicobacter pylori/enzymology , Histidine/metabolism , Histidine/chemistry , Adenylosuccinate Synthase/metabolism , Adenylosuccinate Synthase/chemistry , Adenylosuccinate Synthase/genetics , Kinetics , Circular Dichroism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , X-Ray Diffraction
9.
Sci Rep ; 14(1): 15697, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977739

ABSTRACT

The analysis of recombinant proteins in complex solutions is often accomplished with tag-specific antibodies in western blots. Recently, I introduced an antibody-free alternative wherein tagged proteins are visualized directly within polyacrylamide gels. For this, I used the protein ligase Connectase to selectively attach fluorophores to target proteins possessing an N-terminal recognition sequence. In this study, I extend this methodology to encompass the detection and quantification of C-terminally tagged proteins. Similar to the N-terminal labeling method, this adapted procedure offers increased speed, heightened sensitivity, and an improved signal-to-noise ratio when compared to western blots. It also eliminates the need for sample-specific optimization, enables more consistent and precise quantifications, and uses freely available reagents. This study broadens the applicability of in-gel fluorescence detection methods and thereby facilitates research on recombinant proteins.


Subject(s)
Recombinant Proteins , Recombinant Proteins/metabolism , Recombinant Proteins/chemistry , Fluorescence , Fluorescent Dyes/chemistry , Electrophoresis, Polyacrylamide Gel/methods , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Humans
10.
Appl Microbiol Biotechnol ; 108(1): 424, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037584

ABSTRACT

Leptospirosis, a neglected zoonotic disease, is caused by pathogenic spirochetes belonging to the genus Leptospira and has one of the highest morbidity and mortality rates worldwide. Vaccination stands out as one of the most effective preventive measures for susceptible populations. Within the outer membrane of Leptospira spp., we find the LIC12287, LIC11711, and LIC13259 lipoproteins. These are of interest due to their surface location and potential immunogenicity. Thorough examination revealed the conservation of these proteins among pathogenic Leptospira spp.; we mapped the distribution of T- and B-cell epitopes along their sequences and assessed the 3D structures of each protein. This information aided in selecting immunodominant regions for the development of a chimeric protein. Through gene synthesis, we successfully constructed a chimeric protein, which was subsequently expressed, purified, and characterized. Hamsters were immunized with the chimeric lipoprotein, formulated with adjuvants aluminum hydroxide, EMULSIGEN®-D, Sigma Adjuvant System®, and Montanide™ ISA206VG. Another group was vaccinated with an inactivated Escherichia coli bacterin expressing the chimeric protein. Following vaccination, hamsters were challenged with a virulent L. interrogans strain. Our evaluation of the humoral immune response revealed the production of IgG antibodies, detectable 28 days after the second dose, in contrast to pre-immune samples and control groups. This demonstrates the potential of the chimeric protein to elicit a robust humoral immune response; however, no protection against challenge was achieved. While this study provides valuable insights into the subject, further research is warranted to identify protective antigens that could be utilized in the development of a leptospirosis vaccine. KEY POINTS: • Several T- and B-cell epitopes were identified in all the three proteins. • Four different adjuvants were used in vaccine formulations. • Immunization stimulated significant levels of IgG2/3 in vaccinated animals.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Leptospirosis , Lipoproteins , Animals , Leptospirosis/prevention & control , Leptospirosis/immunology , Lipoproteins/immunology , Lipoproteins/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cricetinae , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Leptospira interrogans/immunology , Leptospira interrogans/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/genetics , Vaccination , Immunity, Humoral , Leptospira/immunology , Leptospira/genetics , Immunogenicity, Vaccine
11.
Front Immunol ; 15: 1392043, 2024.
Article in English | MEDLINE | ID: mdl-38962015

ABSTRACT

In the Americas, P. vivax is the predominant causative species of malaria, a debilitating and economically significant disease. Due to the complexity of the malaria parasite life cycle, a vaccine formulation with multiple antigens expressed in various parasite stages may represent an effective approach. Based on this, we previously designed and constructed a chimeric recombinant protein, PvRMC-1, composed by PvCyRPA, PvCelTOS, and Pvs25 epitopes. This chimeric protein was strongly recognized by naturally acquired antibodies from exposed population in the Brazilian Amazon. However, there was no investigation about the induced immune response of PvRMC-1. Therefore, in this work, we evaluated the immunogenicity of this chimeric antigen formulated in three distinct adjuvants: Stimune, AddaVax or Aluminum hydroxide (Al(OH)3) in BALB/c mice. Our results suggested that the chimeric protein PvRMC-1 were capable to generate humoral and cellular responses across all three formulations. Antibodies recognized full-length PvRMC-1 and linear B-cell epitopes from PvCyRPA, PvCelTOS, and Pvs25 individually. Moreover, mice's splenocytes were activated, producing IFN-γ in response to PvCelTOS and PvCyRPA peptide epitopes, affirming T-cell epitopes in the antigen. While aluminum hydroxide showed notable cellular response, Stimune and Addavax induced a more comprehensive immune response, encompassing both cellular and humoral components. Thus, our findings indicate that PvRMC-1 would be a promising multistage vaccine candidate that could advance to further preclinical studies.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Malaria, Vivax , Mice, Inbred BALB C , Plasmodium vivax , Protozoan Proteins , Animals , Plasmodium vivax/immunology , Plasmodium vivax/genetics , Mice , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Female , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Disease Models, Animal , Adjuvants, Immunologic , Immunogenicity, Vaccine , Antigens, Surface
12.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000180

ABSTRACT

The antimicrobial peptide LRGG (LLRLLRRGGRRLLRLL-NH2) was designed and chemically synthesized in a study conducted by Jia et al. Gram-negative bacteria were found to be sensitive to LRGG and exhibited a high therapeutic index. Genetic engineering methods were used to create the prokaryotic fusion expression vector pQE-GFP-LRGG, and the resulting corresponding fusion protein GFP-LRGG was subsequently expressed and purified. The precursor GFP was then removed by TEV proteolysis, and pure LRGG was obtained after another round of purification and endotoxin removal. The prokaryotic-expressed antimicrobial peptide LRGG displays a broad-spectrum antibacterial effect on Gram-negative bacteria, and its minimum inhibitory activity (MIC) against Escherichia coli can reach 2 µg/mL. Compared to the chemically synthesized LRGG, the prokaryotic-expressed LRGG exhibits similar temperature, pH, salt ion, serum stability, and cell selectivity. Furthermore, prokaryotic-expressed LRGG showed excellent therapeutic effects in both the infection model of cell selectivity and no embryotoxicity in a Galleria mellonella infection model. The mechanism by which LRGG causes bacterial death was found to be the disruption of the Gram-negative cell membrane.


Subject(s)
Antimicrobial Peptides , Microbial Sensitivity Tests , Animals , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/genetics , Antimicrobial Peptides/metabolism , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gram-Negative Bacteria/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/genetics , Humans
13.
J Biotechnol ; 392: 180-189, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39038661

ABSTRACT

Sialylation during N-glycosylation plays an important role in the half-life of therapeutic glycoproteins in vivo and has sparked interest in the production of therapeutic proteins using recombinant Chinese hamster ovary (rCHO) cells. To improve the sialylation of therapeutic proteins, we examined the effect of sialyllactose supplementation on sialylation of Fc-fusion glycoproteins produced in rCHO cells. Two enzymatically-synthesized sialyllactoses, 3'-sialyllactose (3'-SL) and 6'-sialyllactose (6'-SL), were administered separately to two rCHO cell lines producing the same Fc-fusion glycoprotein derived from DUKX-B11 and DG44, respectively. Two sialyllactoses successfully increased sialylation of Fc-fusion glycoprotein in both cell lines, as evidenced by isoform distribution, sialylated N-glycan formation, and sialic acid content. Increased sialylation by adding sialyllactose was likely the result of increased amount of intracellular CMP-sialic acid (CMP-SA), the direct nucleotide sugar for sialylation. Furthermore, the degree of sialylation enhanced by sialyllactoses was slightly effective or nearly similar compared with the addition of N-acetylmannosamine (ManNAc), a representative nucleotide sugar precursor, to increase sialylation of glycoproteins. The effectiveness of sialyllactose was also confirmed using three commercially available CHO cell culture media. Taken together, these results suggest that enzymatically-synthesized sialyllactose represents a promising candidate for culture media supplementation to increase sialylation of glycoproteins in rCHO cell culture.


Subject(s)
Cricetulus , Immunoglobulin Fc Fragments , Lactose , Animals , CHO Cells , Lactose/analogs & derivatives , Lactose/metabolism , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Cricetinae , Glycosylation , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Glycoproteins/metabolism , Glycoproteins/genetics , Culture Media/chemistry , Sialic Acids/metabolism , N-Acetylneuraminic Acid/metabolism , Oligosaccharides
14.
Biomacromolecules ; 25(8): 4639-4662, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39074364

ABSTRACT

Proteins implement many useful functions, including binding ligands with unparalleled affinity and specificity, catalyzing stereospecific chemical reactions, and directing cell behavior. Incorporating proteins into materials has the potential to imbue devices with these desirable traits. This review highlights recent advances in creating active materials by genetically fusing a self-assembling protein to a functional protein. These fusion proteins form materials while retaining the function of interest. Key advantages of this approach include elimination of a separate functionalization step during materials synthesis, uniform and dense coverage of the material by the functional protein, and stabilization of the functional protein. This review focuses on macroscale materials and discusses (i) multiple strategies for successful protein fusion design, (ii) successes and limitations of the protein fusion approach, (iii) engineering solutions to bypass any limitations, (iv) applications of protein fusion materials, including tissue engineering, drug delivery, enzyme immobilization, electronics, and biosensing, and (v) opportunities to further develop this useful technique.


Subject(s)
Biocompatible Materials , Recombinant Fusion Proteins , Biocompatible Materials/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Humans , Protein Engineering/methods , Animals , Tissue Engineering/methods , Proteins/chemistry , Proteins/genetics , Drug Delivery Systems/methods
15.
PeerJ ; 12: e17806, 2024.
Article in English | MEDLINE | ID: mdl-39035165

ABSTRACT

Epidermal growth factor (EGF) protein is a crucial biomolecule involved in regulating cell growth, proliferation, migration and differentiation, which is used in various therapeutic applications, such as wound healing and tissue regeneration. The production of recombinant EGF is essential for studying its biological function and for its clinical translation. However, EGF protein expressed in prokaryotic cells often occurs in inclusion bodies, and co-expression with soluble tag protein is an effective method to prepare recombinant EGF. In this study, we expressed recombinant human EGF (rhEGF) fused to a HaloTag (Halo-rhEGF) and a large portion of Halo-rhEGF was found in the soluble fraction. Cell growth assay showed that the purified Halo-rhEGF protein could promote the proliferation of fibroblasts (NIH 3T3) and epithelial cells (HaCaT), and significantly increased their viability. Phosphorylation of the intracellular signaling proteins, ERK1/2 and c-Jun, was stimulated by treatment with Halo-rhEGF and the expression levels of proteins regulating cell proliferation were significantly increased. RNA sequencing analysis revealed that rhEGF could increase the transcription of genes enriched in ribosome generation and cell proliferation. Moreover, Halo-rhEGF can be labelled by HaloTag ligand for fluorescence imaging and can be slowly released in tissue repair by binding to anion biomaterials. In conclusion, HaloTag is an efficient fusion tag for rhEGF protein expression, purification and controlled release, and Halo-rhEGF can promote the proliferation and viability of epithelial and fibroblast cells.


Subject(s)
Cell Proliferation , Epidermal Growth Factor , Humans , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/genetics , Cell Proliferation/drug effects , Mice , Animals , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , NIH 3T3 Cells , Cell Survival/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/metabolism
16.
Curr Protein Pept Sci ; 25(7): 567-576, 2024.
Article in English | MEDLINE | ID: mdl-39044556

ABSTRACT

BACKGROUND: Vascular Endothelial Growth Factor Receptors (VEGFR1 and VEGFR2) are tyrosine kinase receptors expressed on endothelial cells and tumor vessels and play an important role in angiogenesis. In this study, three repeats of VEGFR1 and VEGFR2 binding peptide (VGB3) were genetically fused to the truncated diphtheria toxin (TDT), and its in vitro activity was evaluated. METHODS: The recombinant construct (TDT-triVGB3) was expressed in bacteria cells and purified with nickel affinity chromatography. The binding capacity and affinity of TDT-triVGB3 were evaluated using the enzyme-linked immunosorbent assay. The inhibitory activity of TDT-triVGB3 on viability, migration, and tube formation of human endothelial cells was evaluated using MTT, migration, and tube formation assays. RESULTS: TDT-triVGB3 selectively detected VEGFR1 and VEGFR2 with high affinity in an enzyme- linked immunosorbent assay and significantly inhibited viability, migration, and tube formation of human endothelial cells. CONCLUSION: The developed TDT-triVGB3 is potentially a novel agent for targeting VEGFR1/ VEGFR2 over-expressing cancer cells.


Subject(s)
Angiogenesis Inhibitors , Cell Movement , Diphtheria Toxin , Human Umbilical Vein Endothelial Cells , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factor Receptor-2 , Humans , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Diphtheria Toxin/genetics , Diphtheria Toxin/pharmacology , Diphtheria Toxin/metabolism , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/chemistry , Cell Survival/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/drug therapy , Gene Expression , Endothelial Cells/metabolism , Endothelial Cells/drug effects
17.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2150-2161, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044581

ABSTRACT

This study aims to develop an effective bivalent subunit vaccine that is promising to prevent both porcine deltacoronavirus (PDCoV) and porcine epidemic diarrhea virus (PEDV). The receptor-binding domains (RBDs) of PDCoV and PEDV were fused and cloned into the eukaryotic expression vector pCDNA3.1(+). The fusion protein PDCoV-RBD-PEDV-RBD (pdRBD-peRBD) was expressed by the ExpiCHOTM expression system and purified. Mice were immunized with the fusion protein at three different doses (10, 20, and 30 µg). The humoral immune response and cellular immune response induced by the fusion protein were evaluated by ELISA and flow cytometry. The neutralization titers of the serum of immunized mice against PDCoV and PEDV were determined by the microneutralization test. The results showed that high levels of IgG antibodies were induced in the three different dose groups after booster immunization, and there was no significant difference in the antibody level between different dose groups, indicating that the immunization dose of 10 µg could achieve the fine immune effect. The results of flow cytometry showed that the immunization groups demonstrated increased proportion of CD3+CD4+ T cells and decreased proportion of CD3+CD8+ T cells, which was consistent with the expectation about the humoral immune response induced by the subunit vaccine. At the same time, the levels of interleukin (IL)-2, IL-4, and interferon (IFN)-γ in the serum were determined. The results showed that the fusion protein induced both humoral immune effect and cellular immune response. The results of the neutralization test showed that the antibody induced by 10 µg fusion protein neutralized both PDCoV and PEDV in vitro, with the titers of 1:179.25 and 1:141.21, respectively. The above results suggested that the pdRBD-peRBD could induce a high level of humoral immune response at a dose of 10 µg, and the induced antibody could neutralize both PDCoV and PEDV. Therefore, the fusion protein pdRBD-peRBD is expected to be an effective subunit vaccine that can simultaneously prevent PDCoV and PEDV.


Subject(s)
Antibodies, Viral , Coronavirus Infections , Porcine epidemic diarrhea virus , Recombinant Fusion Proteins , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/genetics , Mice , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Deltacoronavirus/immunology , Deltacoronavirus/genetics , Swine Diseases/prevention & control , Swine Diseases/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Protein Domains , Immunogenicity, Vaccine , Immunity, Humoral
18.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2258-2269, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044589

ABSTRACT

This study aims to prepare bacterial outer membrane vesicles (OMVs) with anti-glypican-3 (GPC3) single-chain antibody and analyze their targeting effects on Hep G2 hepatocellular carcinoma (HCC) cells and tissue. The recombinant plasmid pET28a-Hbp-hGC 33-scFv was constructed by ligating Hbp-hGC 33-scFv to pET28a. Western blotting was employed to determine the prokaryotic expression of the fusion protein Hbp-hGC 33-scFv, on the basis of which the optimal induction conditions were determined. Hbp-hGC 33-OMVs secreted from the recombinant expressing strains were collected by ultrafiltration concentration and then characterized. The localization of Hbp-hGC 33-scFv in bacteria and Hbp-hGC 33-OMVs was analyzed by immune electron microscopy. The binding of Hbp-hGC 33-scFv to Hep G2 cells was observed by immunofluorescence. The Hep G2 tumor-bearing mouse model was established, and the targeted retention of Hbp-hGC 33-OMVs in the tumor site of mice was observed by a fluorescence imaging system in vivo. The results showed that the actual molecular weight of the fusion protein was 175.3 kDa, and the optimal induction conditions were as follows: OD600=0.5, IPTG added at a final concentration of 0.5 mmol/L, and overnight induction at 16 ℃. The prepared Hbp-hGC 33-OMVs were irregular spherical structures with an average particle size of (112.3±4.6) nm, expressing OmpC, OmpA, and the fusion protein Hbp-hGC 33-scFv. The Hbp-hGC 33-OMVs prepared in this study demonstrated stronger ability of binding to Hep G2 cells than the wild-type OMVs (P=0.008). All the data indicated that Hbp-hGC 33-OMVs with anti-GPC3 single-chain antibody were successfully prepared and could be used for research on the targeted therapy of hepatocellular carcinoma.


Subject(s)
Bacterial Outer Membrane , Carcinoma, Hepatocellular , Glypicans , Liver Neoplasms , Single-Chain Antibodies , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/chemistry , Animals , Mice , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Bacterial Outer Membrane/metabolism , Bacterial Outer Membrane/immunology , Hep G2 Cells , Glypicans/immunology , Glypicans/metabolism , Glypicans/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Drug Delivery Systems , Mice, Nude
19.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2282-2293, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39044591

ABSTRACT

The ubiquitin/proteasome system (UPS) plays a crucial role in maintaining cellular protein homeostasis. The catalytic activity of proteasome in the UPS is regulated by ß1 (PSMB6), ß2 (PSMB7), and ß5 (PSMB5) subunits. Interferon (IFN)-γ, tumor necrosis factor (TNF)-α, inflammation, and oxidative stress can induce the replacement of ß1, ß2, and ß5 with their respective immuno-subunits ß1i (PSMB9), ß2i (PSMB10), and ß5i (PSMB8), which can be assembled into the immunoproteasome. Compared with the standard proteasome, the immunoproteasome exerts enhanced regulatory effects on immune responses, such as processing and presenting MHC class Ⅰ antigens, production of pro-inflammatory cytokines, and T cell differentiation and proliferation. Abnormal aggregation of immunoproteasomes can cause neurodegenerative diseases like Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis. To explore the function of PSMB9 after bacterial infection, we constructed a lentivirus plasmid overexpressing PSMB9-eGFP-His and transfected the plasmid into HEK293T cells for packaging by using a triple-plasmid system in this study. After screening with puromycin, we obtained a stable human leukemia monocytic THP-1 cell line expressing the fusion protein of PSMB9. Western blotting (WB) and fluorescence microscopy verified the expression of the fusion protein in the stable THP-1 cells. Quantitative PCR (qPCR) was employed to measure the copies of PSMB9-eGFP in THP-1 cells. Immunofluorescence results found that eGFP-His did not affect the subcellular localization of PSMB9. The purification with nickel affinity chromatography confirmed that the fusion protein could be assembled into the 20S immunoproteasome and exhibited cleaving activity for fluorescent peptide substrates. These results indicated that the PSMB9-eGFP fusion gene was integrated into the chromosome, and could be stably expressed in the constructed THP-1 cell line. This cell line can be utilized for the research on subcellular localization, dynamic expression, and activity of PSMB9 in live cells at different infection conditions and disease stages. It also provides a model for the stable cell lines construction of other immunoproteasome subunits PSMB8 and PSMB10.


Subject(s)
Green Fluorescent Proteins , Proteasome Endopeptidase Complex , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , THP-1 Cells , Lentivirus/genetics , Recombinant Fusion Proteins/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism
20.
PLoS Negl Trop Dis ; 18(7): e0012320, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39008519

ABSTRACT

BACKGROUND: The sensitivity of parasitological and molecular methods is unsatisfactory for the diagnosis of strongyloidiasis, and serological techniques are remaining as the most effective diagnostic approach. The present study aimed to design and produce a chimeric recombinant antigen from Strongyloides stercoralis immunoreactive antigen (SsIR) and Ss1a antigens, using immune-informatics approaches, and evaluated its diagnostic performance in an ELISA system for the diagnosis of human strongyloidiasis. METHODOLOGY/PRINCIPAL FINDINGS: The coding sequences for SsIR and Ss1a were selected from GenBank and were gene-optimized. Using bioinformatics analysis, the regions with the highest antigenicity that did not overlap with other parasite antigens were selected. The chimeric recombinant antigen SsIR- Ss1a, was constructed. The solubility and physicochemical properties of the designed construct were analyzed and its tertiary structures were built and evaluated. The construct was expressed into the pET-23a (+) expression vector and the optimized DNA sequences of SsIR-Ss1a (873 bp) were cloned into competent E. coli DH5α cells. Diagnostic performances of the produced recombinant antigen, along with a commercial kit were evaluated in an indirect ELISA system, using a panel of sera from strongyloidiasis patients and controls. The physicochemical and bioinformatics evaluations revealed that the designed chimeric construct is soluble, has a molecular with of 35 KDa, and is antigenic. Western blotting confirmed the immunoreactivity of the produced chimeric recombinant antigen with the sera of strongyloidiasis patients. The sensitivity and specificity of the indirect ELISA system, using the produced SsIR-Ss1a chimeric antigen, were found to be 93.94% (95% CI, 0.803 to 0.989) and 97.22% (95% CI, 0.921 to 0.992) respectively. CONCLUSIONS/SIGNIFICANCE: The preliminary findings of this study suggest that the produced SsIR-Ss1a chimeric antigen shows promise in the diagnosis of human strongyloidiasis. However, these results are based on a limited panel of samples, and further research with a larger sample size is necessary to confirm its accuracy. The construct has potential as an antigen in the ELISA system for the serological diagnosis of this neglected parasitic infection, but additional validation is required.


Subject(s)
Antigens, Helminth , Enzyme-Linked Immunosorbent Assay , Sensitivity and Specificity , Serologic Tests , Strongyloides stercoralis , Strongyloidiasis , Humans , Strongyloidiasis/diagnosis , Strongyloidiasis/immunology , Animals , Serologic Tests/methods , Enzyme-Linked Immunosorbent Assay/methods , Strongyloides stercoralis/immunology , Strongyloides stercoralis/genetics , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Antibodies, Helminth/blood , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Gene Expression
SELECTION OF CITATIONS
SEARCH DETAIL