Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(13): e23759, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38949635

ABSTRACT

The epidermal growth factor receptor (EGFR) is an important target for cancer therapies. Many head and neck cancer (HNC) cells have been reported to overexpress EGFR; therefore, anti-EGFR therapies have been attempted in patients with HNC. However, its clinical efficacy is limited owing to the development of drug resistance. In this study, we developed an EGFR-targeting immunotoxin consisting of a clinically proven anti-EGFR IgG (cetuximab; CTX) and a toxin fragment (LR-LO10) derived from Pseudomonas exotoxin A (PE) using a novel site-specific conjugation technology (peptide-directed photo-crosslinking reaction), as an alternative option. The immunotoxin (CTX-LR-LO10) showed specific binding to EGFR and properties of a typical IgG, such as stability, interactions with receptors of immune cells, and pharmacokinetics, and inhibited protein synthesis via modification of elongation factor-2. Treatment of EGFR-positive HNC cells with the immunotoxin resulted in apoptotic cell death and the inhibition of cell migration and invasion. The efficacy of CTX-LR-LO10 was evaluated in xenograft mouse models, and the immunotoxin exhibited much stronger tumor suppression than CTX or LR-LO10. Transcriptome analyses revealed that the immunotoxins elicited immune responses and altered the expression of genes related to its mechanisms of action. These results support the notion that CTX-LR-LO10 may serve as a new therapeutic agent targeting EGFR-positive cancers.


Subject(s)
ADP Ribose Transferases , ErbB Receptors , Exotoxins , Head and Neck Neoplasms , Immunoglobulin G , Immunotoxins , Pseudomonas aeruginosa Exotoxin A , Virulence Factors , Humans , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , ErbB Receptors/immunology , Animals , Immunotoxins/pharmacology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Mice , Immunoglobulin G/pharmacology , Cell Line, Tumor , Exotoxins/pharmacology , Xenograft Model Antitumor Assays , Cetuximab/pharmacology , Mice, Nude , Bacterial Toxins , Apoptosis/drug effects , Mice, Inbred BALB C , Female , Cell Movement/drug effects , Antineoplastic Agents/pharmacology
2.
Sci Rep ; 13(1): 18275, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880350

ABSTRACT

Complement-dependent cytotoxicity (CDC), which eliminates aberrant target cells through the assembly and complex formation of serum complement molecules, is one of the major effector functions of anticancer therapeutic antibodies. In this study, we discovered that breaking the symmetry of natural immunoglobulin G (IgG) antibodies significantly increased the CDC activity of anti-CD20 antibodies. In addition, the expression of CD55 (a checkpoint inhibitor in the CDC cascade) was significantly increased in a rituximab-resistant cell line generated in-house, suggesting that CD55 overexpression might be a mechanism by which cancer cells acquire rituximab resistance. Based on these findings, we developed an asymmetric bispecific antibody (SBU-CD55 × CD20) that simultaneously targets both CD55 and CD20 to effectively eliminate rituximab-resistant cancer cells. In various cancer cell lines, including rituximab-resistant lymphoma cells, the SBU-CD55 × CD20 antibody showed significantly higher CDC activity than either anti-CD20 IgG antibody alone or a combination of anti-CD20 IgG antibody and anti-CD55 IgG antibody. Furthermore, the asymmetric bispecific antibody (SBU-CD55 × CD20) exhibited significantly higher CDC activity against rituximab-resistant cancer cells compared to other bispecific antibodies with symmetric features. These results demonstrate that enhancing CDC with an asymmetric CD55-binding bispecific antibody could be a new strategy for developing therapeutics to treat patients with relapsed or refractory cancers.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Humans , Rituximab/pharmacology , Immunoglobulin G , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20 , CD55 Antigens/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antibodies, Bispecific/pharmacology , Cell Line, Tumor , Antibody-Dependent Cell Cytotoxicity
3.
Mol Pharm ; 20(4): 2170-2180, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36897183

ABSTRACT

Although therapeutic immunoglobulin G (IgG) antibodies that regulate the activity of immune checkpoints bring innovation to the field of immuno-oncology, they are still limited in their efficiency to infiltrate the tumor microenvironment due to their large molecular size (150 kDa) and the necessity of additional engineering works to ablate effector functions for antibodies targeting immune cells. To address these issues, the human PD-1 (hPD-1) ectodomain, a small protein moiety of 14-17 kDa, has been considered as a therapeutic agent. Here, we used bacterial display-based high-throughput directed evolution to successfully isolate glycan-controlled (aglycosylated or only single-N-linked glycosylated) human PD-1 variants exhibiting over 1000-fold increased hPD-L1 binding affinity compared to that of wild-type hPD-1. The resulting hPD-1 variants, aglycosylated JYQ12 and JYQ12-2 with a single-N-linked glycan chain, showed exceptionally high binding affinity to hPD-L1 and very high affinity to both hPD-L2 and mPD-L1. Moreover, the JYQ12-2 efficiently potentiated the proliferation of human T cells. hPD-1 variants with significantly improved binding affinities for hPD-1 ligands could be used as effective therapeutics or diagnostics that can be differentiated from large-sized IgG antibody-based molecules.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , T-Lymphocytes/metabolism , Programmed Cell Death 1 Receptor/metabolism , Neoplasms/metabolism , Tumor Microenvironment
4.
Biotechnol Lett ; 45(5-6): 589-600, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36971774

ABSTRACT

OBJECTIVES: S100A8 is highly expressed in several inflammatory and oncological conditions. To address the current lack of a reliable and sensitive detection method for S100A8, we generated a monoclonal antibody with a high binding affinity to human S100A8 to enable early disease diagnosis. RESULTS: A soluble recombinant S100A8 protein with a high yield and purity was produced using Escherichia coli. Next, mice were immunized with recombinant S100A8 to obtain anti-human S100A8 monoclonal antibodies using hybridoma technology. Lastly, the high binding activity of the antibody was confirmed and its sequence was identified. CONCLUSIONS: This method, including the production of antigens and antibodies, will be useful for the generation of hybridoma cell lines that produce anti-S100A8 monoclonal antibodies. Moreover, the sequence information of the antibody can be used to develop a recombinant antibody for use in various research and clinical applications.


Subject(s)
Antibodies, Monoclonal , Calgranulin A , Animals , Mice , Antibodies, Monoclonal/chemistry , Hybridomas , Cell Line , Recombinant Proteins/genetics , Biomarkers
5.
Mol Pharm ; 20(2): 1247-1255, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36563318

ABSTRACT

Endothelin receptor A (ETA), a class A G protein-coupled receptor (GPCR), is a promising tumor-associated antigen due to its close association with the progression and metastasis of many types of cancer, such as colorectal, breast, lung, ovarian, and prostate cancer. However, only small-molecule drugs have been developed as ETA antagonists with anticancer effects. In a previous study, we identified an antibody (AG8) with highly selective binding to human ETA through screening of a human naïve immune antibody library. Although both in vitro and in vivo experiments indicated that the identified AG8 had anticancer effects, there is a need for improvement in biochemical and physicochemical properties such as the ETA binding affinity, thermostability, and productivity. In this study, we engineered the framework regions of AG8 and isolated an anti-ETA antibody (MJF1) exhibiting significantly improved thermostability and ETA binding affinity. Subsequently, our previously isolated PFc29, an Fc variant with an enhanced pH-dependent human FcRn binding profile, was introduced to MJF1, and the resulting Fc-engineered anti-ETA antibody (MJF1-PFc29) inhibited the proliferation of tumor cells comparably to MJF1 and showed a 4.2-fold increased serum half-life in human FcRn transgenic mice. Moreover, MJF1-PFc29 elicited higher tumor growth inhibition in colorectal cancer xenograft mice compared to MJF1. Our results demonstrate that the engineered human anti-ETA antibody MJF1-PFc29 has great therapeutic potential and high antitumor potency against various types of cancers including colorectal cancer.


Subject(s)
Colorectal Neoplasms , Protein Engineering , Male , Humans , Mice , Animals , Receptors, Fc/metabolism , Mice, Transgenic , Receptor, Endothelin A , Colorectal Neoplasms/drug therapy
6.
Exp Mol Med ; 54(11): 1850-1861, 2022 11.
Article in English | MEDLINE | ID: mdl-36319752

ABSTRACT

The pH-selective interaction between the immunoglobulin G (IgG) fragment crystallizable region (Fc region) and the neonatal Fc receptor (FcRn) is critical for prolonging the circulating half-lives of IgG molecules through intracellular trafficking and recycling. By using directed evolution, we successfully identified Fc mutations that improve the pH-dependent binding of human FcRn and prolong the serum persistence of a model IgG antibody and an Fc-fusion protein. Strikingly, trastuzumab-PFc29 and aflibercept-PFc29, a model therapeutic IgG antibody and an Fc-fusion protein, respectively, when combined with our engineered Fc (Q311R/M428L), both exhibited significantly higher serum half-lives in human FcRn transgenic mice than their counterparts with wild-type Fc. Moreover, in a cynomolgus monkey model, trastuzumab-PFc29 displayed a superior pharmacokinetic profile to that of both trastuzumab-YTE and trastuzumab-LS, which contain the well-validated serum half-life extension Fcs YTE (M252Y/S254T/T256E) and LS (M428L/N434S), respectively. Furthermore, the introduction of two identified mutations of PFc29 (Q311R/M428L) into the model antibodies enhanced both complement-dependent cytotoxicity and antibody-dependent cell-mediated cytotoxicity activity, which are triggered by the association between IgG Fc and Fc binding ligands and are critical for clearing cancer cells. In addition, the effector functions could be turned off by combining the two mutations of PFc29 with effector function-silencing mutations, but the antibodies maintained their excellent pH-dependent human FcRn binding profile. We expect our Fc variants to be an excellent tool for enhancing the pharmacokinetic profiles and potencies of various therapeutic antibodies and Fc-fusion proteins.


Subject(s)
Histocompatibility Antigens Class I , Immunoglobulin G , Mice , Animals , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Macaca fascicularis/metabolism , Half-Life , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Mice, Transgenic , Mutation , Trastuzumab/therapeutic use , Trastuzumab/genetics
7.
Sci Rep ; 12(1): 19030, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36347901

ABSTRACT

Cancer therapy using immune checkpoint inhibitor antibodies has markedly shifted the paradigm of cancer treatment. However, methods completely eliminating the effector function of these signal-regulating antibodies is urgently required. The heterogeneity of glycan chains in antibodies limits their use as therapeutic agents due to their variability; thus, the development of uniform glycan chains is necessary. Here, we subjected the anti-programmed cell death protein (PD)-1 antibody nivolumab, a representative immune checkpoint inhibitor, to GlycoDelete (GD) engineering to remove the antibody-dependent cellular cytotoxicity (ADCC) of the antibody, leaving only one glycan in the Fc. Glyco-engineered CHO cells were prepared by overexpressing endo-ß-N-acetyl-glucosaminidase (Endo T) in CHO cells, in which N-acetyl-glucosaminyl-transferase I was knocked out using Cas9. GD IgG1 nivolumab and GD IgG4 nivolumab were produced using GD CHO cells, and glycan removal was confirmed using mass spectrometry. Target binding and PD-1 inhibition was not altered; however, ADCC decreased. Furthermore, the IgG4 form, determined to be the most suitable form of GD nivolumab, was produced in a plant GD system. The plant GD nivolumab also reduced ADCC without affecting PD-1 inhibitory function. Thus, CHO and plant GD platforms can be used to improve signal-regulating antibodies by reducing their effector function.


Subject(s)
Immunoglobulin Fc Fragments , Nivolumab , Cricetinae , Animals , Cricetulus , Immunoglobulin Fc Fragments/metabolism , Programmed Cell Death 1 Receptor , Immune Checkpoint Inhibitors , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin G , Polysaccharides/metabolism , Receptors, IgG/metabolism
8.
Front Chem ; 10: 839636, 2022.
Article in English | MEDLINE | ID: mdl-35295971

ABSTRACT

Non-canonical amino acids (ncAAs) have been utilized as an invaluable tool for modulating the active site of the enzymes, probing the complex enzyme mechanisms, improving catalytic activity, and designing new to nature enzymes. Here, we report site-specific incorporation of p-benzoyl phenylalanine (pBpA) to engineer (R)-amine transaminase previously created from d-amino acid aminotransferase scaffold. Replacement of the single Phe88 residue at the active site with pBpA exhibits a significant 15-fold and 8-fold enhancement in activity for 1-phenylpropan-1-amine and benzaldehyde, respectively. Reshaping of the enzyme's active site afforded an another variant F86A/F88pBpA, with 30% higher thermostability at 55°C without affecting parent enzyme activity. Moreover, various racemic amines were successfully resolved by transaminase variants into (S)-amines with excellent conversions (∼50%) and enantiomeric excess (>99%) using pyruvate as an amino acceptor. Additionally, kinetic resolution of the 1-phenylpropan-1-amine was performed using benzaldehyde as an amino acceptor, which is cheaper than pyruvate. Our results highlight the utility of ncAAs for designing enzymes with enhanced functionality beyond the limit of 20 canonical amino acids.

9.
Mol Oncol ; 16(6): 1290-1308, 2022 03.
Article in English | MEDLINE | ID: mdl-34407310

ABSTRACT

c-Kit overexpression and activating mutations, which are reported in various cancers, including gastrointestinal stromal tumor (GIST), small-cell lung cancer (SCLC), acute myeloid leukemia, acral melanoma, and systemic mastocytosis (SM), confer resistance to tyrosine kinase inhibitors (TKIs). To overcome TKI resistance, an anti-c-Kit antibody-drug conjugate was developed in this study to treat wild-type and mutant c-Kit-positive cancers. NN2101, a fully human IgG1, was conjugated to DM1, a microtubule inhibitor, through N-succinimidyl-4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) (to give NN2101-DM1). The antitumor activity of NN2101-DM1 was evaluated in vitro and in vivo using various cancer cell lines. NN2101-DM1 exhibited potent growth-inhibitory activities against c-Kit-positive cancer cell lines. In a mouse xenograft model, NN2101-DM1 exhibited potent growth-inhibitory activities against imatinib-resistant GIST and SM cells. In addition, NN2101-DM1 exhibited a significantly higher anti-cancer effect than carboplatin/etoposide against SCLC cells where c-Kit does not mediate cancer pathogenesis. Furthermore, the combination of NN2101-DM1 with imatinib in imatinib-sensitive GIST cells induced complete remission compared with treatment with NN2101-DM1 or imatinib alone in mouse xenograft models. These results suggest that NN2101-DM1 is a potential therapeutic agent for wild-type and mutant c-Kit-positive cancers.


Subject(s)
Gastrointestinal Stromal Tumors , Immunoconjugates , Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/drug therapy , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/genetics
10.
Proteomics ; 22(1-2): e2100171, 2022 01.
Article in English | MEDLINE | ID: mdl-34561969

ABSTRACT

Human leukocyte antigen (HLA) class I has more than 18,000 alleles, each of which binds to a set of unique peptides from the cellular degradome. Deciphering the interaction between antigenic peptides and HLA proteins is crucial for understanding immune responses in autoimmune diseases and cancer. In this study, we aimed to characterize the peptidome that binds to HLA-A*33:03, which is one of the most prevalent HLA-A alleles in the Northeast Asian population, but poorly studied. For this purpose, we analyzed the HLA-A*33:03 monoallelic B cell line using immunoprecipitation of HLA-A and peptide complexes, followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). In this study, we identified 5731 unique peptides that were associated with HLA A*33:03, and experimentally validated the affinity of 40 peptides for HLA-A*33:03 and their stability in HLA A*33:03-peptides complexes. To our knowledge, this study represents the largest dataset of peptides associated with HLA-A*33:03. Also, this is the first study in which HLA A*33:03-associated peptides were experimentally validated.


Subject(s)
HLA-A Antigens , Tandem Mass Spectrometry , Chromatography, Liquid , Epitopes , Humans , Immunoprecipitation
11.
Front Chem ; 9: 772648, 2021.
Article in English | MEDLINE | ID: mdl-34765589

ABSTRACT

In the last two decades, methods to incorporate non-canonical amino acids (ncAAs) into specific positions of a protein have advanced significantly; these methods have become general tools for engineering proteins. However, almost all these methods depend on the translation elongation process, and strategies leveraging the initiation process have rarely been reported. The incorporation of a ncAA specifically at the translation initiation site enables the installation of reactive groups for modification at the N-termini of proteins, which are attractive positions for introducing abiological groups with minimal structural perturbations. In this study, we attempted to engineer an orthogonal protein translation initiation system. Introduction of the identity elements of Escherichia coli initiator tRNA converted an engineered Methanococcus jannaschii tRNATyr into an initiator tRNA. The engineered tRNA enabled the site-specific incorporation of O-propargyl-l-tyrosine (OpgY) into the amber (TAG) codon at the translation initiation position but was inactive toward the elongational TAG codon. Misincorporation of Gln was detected, and the engineered system was demonstrated only with OpgY. We expect further engineering of the initiator tRNA for improved activity and specificity to generate an orthogonal translation initiation system.

12.
Exp Mol Med ; 53(9): 1437-1448, 2021 09.
Article in English | MEDLINE | ID: mdl-34588605

ABSTRACT

Endothelin receptor A (ETA), a class A G-protein-coupled receptor (GPCR), is involved in the progression and metastasis of colorectal, breast, lung, ovarian, and prostate cancer. We overexpressed and purified human endothelin receptor type A in Escherichia coli and reconstituted it with lipid and membrane scaffold proteins to prepare an ETA nanodisc as a functional antigen with a structure similar to that of native GPCR. By screening a human naive immune single-chain variable fragment phage library constructed in-house, we successfully isolated a human anti-ETA antibody (AG8) exhibiting high specificity for ETA in the ß-arrestin Tango assay and effective inhibitory activity against the ET-1-induced signaling cascade via ETA using either a CHO-K1 cell line stably expressing human ETA or HT-29 colorectal cancer cells, in which AG8 exhibited IC50 values of 56 and 51 nM, respectively. In addition, AG8 treatment repressed the transcription of inhibin ßA and reduced the ETA-induced phosphorylation of protein kinase B and extracellular regulated kinase. Furthermore, tumor growth was effectively inhibited by AG8 in a colorectal cancer mouse xenograft model. The human anti-ETA antibody isolated in this study could be used as a potential therapeutic for cancers, including colorectal cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Endothelin A Receptor Antagonists/pharmacology , Receptor, Endothelin A/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antineoplastic Agents, Immunological/chemistry , CHO Cells , Cell Line, Tumor , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelin A Receptor Antagonists/chemistry , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Protein Engineering , Xenograft Model Antitumor Assays
13.
Protein Expr Purif ; 187: 105931, 2021 11.
Article in English | MEDLINE | ID: mdl-34197919

ABSTRACT

We have successfully produced a recombinant human matrix metalloproteinase 9 (hMMP9) antigen with high yield and purity and used it to generate a hybridoma cell-culture-based monoclonal anti-hMMP9 antibody. We selected the most effective antibody for binding antigens and successfully identified its nucleotide sequence. The entire antigen and antibody developmental procedures described herein can be a practical approach for producing large amounts of monoclonal antibodies against hMMP9 and other antigens of interest. Additionally, the nucleotide sequence information of the anti-hMMP9 monoclonal antibody revealed herein will be useful for the generation of recombinant antibodies or antibody fragments against hMMP9.


Subject(s)
Antibodies, Monoclonal/genetics , Matrix Metalloproteinase 9/genetics , Recombinant Proteins/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Base Sequence , Cell Culture Techniques , Gene Expression Regulation , Humans , Hybridomas/cytology , Immunoglobulin Fragments/chemistry , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/immunology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Solubility
14.
Mol Biotechnol ; 63(11): 1016-1029, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34185248

ABSTRACT

Plants are promising drug-production platforms with high economic efficiency, stability, and convenience in mass production. However, studies comparing the equivalency between the original antibodies and those produced in plants are limited. Amino acid sequences that constitute the Fab region of an antibody are diverse, and the post-transcriptional modifications that occur according to these sequences in animals and plants are also highly variable. In this study, rituximab, a blockbuster antibody drug used in the treatment of non-Hodgkin's lymphoma, was produced in Nicotiana benthamiana leaves and Arabidopsis thaliana callus, and was compared to the original rituximab produced in CHO cells. Interestingly, the epitope recognition and antigen-binding abilities of rituximab from N. benthamiana leaves were almost lost. In the case of rituximab produced in A. thaliana callus, the specific binding ability and CD20 capping activity were maintained, but the binding affinity was less than 50% of that of original rituximab from CHO cells. These results suggest that different plant species exhibit different binding affinities. Accordingly, in addition to the differences in PTMs between mammals and plants, the differences between the species must also be considered in the process of producing antibodies in plants.


Subject(s)
Antigens, CD20/metabolism , Arabidopsis/metabolism , Nicotiana/metabolism , Plant Leaves/chemistry , Rituximab/metabolism , Animals , Antibody Affinity , Antigens, CD20/chemistry , Antineoplastic Agents, Immunological/isolation & purification , Antineoplastic Agents, Immunological/metabolism , Arabidopsis/genetics , Cricetinae , Humans , Plant Leaves/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Rituximab/biosynthesis , Rituximab/genetics , Rituximab/isolation & purification , Nicotiana/genetics
15.
Chem Sci ; 11(31): 8288-8294, 2020 Jul 24.
Article in English | MEDLINE | ID: mdl-34094182

ABSTRACT

Two-phenoxy walled calix[4]pyrroles 1 and 2 strapped with small rigid linkers containing pyridine and benzene, respectively, have been synthesized. 1H NMR spectroscopic analyses carried out in CDCl3 revealed that both of receptors 1 and 2 recognize only F- and HCO3 - among various test anions with high preference for HCO3 - (as the tetraethylammonium, TEA+ salt) relative to F- (as the TBA+ salt). The bound HCO3 - anion was completely released out of the receptors upon the addition of F- (as the tetrabutylammonium, TBA+ salt) as a result of significantly enhanced affinities and selectivities of the receptors for F- once converted to the TEAHCO3 complexes. Consequently, relatively stable TEAF complexes of receptors 1 and 2 were formed via anion metathesis occurring within the receptor cavities. By contrast, the direct addition of TEAF to receptors 1 and 2 produces different complexation products initially, although eventually the same TEAF complexes are produced as via sequential TEAHCO3 and TBAF addition. These findings are rationalized in terms of the formation of different ion pair complexes involving interactions both inside and outside of the core receptor framework.

16.
RSC Adv ; 10(40): 23792-23800, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-35517354

ABSTRACT

The halophilic bacterium Pseudoalteromonas phenolica is well known as a promising candidate that enables the recycling of organic wastes at high salinity. However, for industrial applications of P. phenolica further research is required to explore the biological mechanism for maximizing the activities and productivities of this bacterium. In this study, we investigated the osmotic stress resistance and specific protease activities of P. phenolica in a normal-salt medium (0.3 M NaCl) and high-salt medium (1 M NaCl) based on intra- and extracellular multi-omics approaches. Proteins related to betaine and proline biosynthesis were increased under high salt stress. The targeted metabolite analysis found that proline was overproduced and accumulated outside the cell at high salinity, and betaine was accumulated in the cell by activation of biosynthesis as well as uptake. In addition, extracellular serine proteases were shown to be upregulated in response to salt stress by the extracellular proteomic analysis. The specific proteolytic activity assay indicated that the activities of serine proteases, useful enzymes for the recycling of organic wastes, were increased remarkably under high salt stress. Our results suggest that betaine and proline are key osmoprotectant metabolites of P. phenolica, and they can be used for the improvement of protease production and P. phenolica activities for the recycling of high-salt organic wastes in the future.

17.
Antibodies (Basel) ; 8(3)2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31544848

ABSTRACT

Since its first report in the Middle East in 2012, the Middle East respiratory syndrome-coronavirus (MERS-CoV) has become a global concern due to the high morbidity and mortality of individuals infected with the virus. Although the majority of MERS-CoV cases have been reported in Saudi Arabia, the overall risk in areas outside the Middle East remains significant as inside Saudi Arabia. Additional pandemics of MERS-CoV are expected, and thus novel tools and reagents for therapy and diagnosis are urgently needed. Here, we used phage display to develop novel monoclonal antibodies (mAbs) that target MERS-CoV. A human Fab phage display library was panned against the S2 subunit of the MERS-CoV spike protein (MERS-S2P), yielding three unique Fabs (S2A3, S2A6, and S2D5). The Fabs had moderate apparent affinities (Half maximal effective concentration (EC50 = 123-421 nM) for MERS-S2P, showed no cross-reactivity to spike proteins from other CoVs, and were non-aggregating and thermostable (Tm = 61.5-80.4 °C). Reformatting the Fabs into IgGs (Immunoglobulin Gs) greatly increased their apparent affinities (KD = 0.17-1.2 nM), presumably due to the effects of avidity. These apparent affinities were notably higher than that of a previously reported anti-MERS-CoV S2 reference mAb (KD = 8.7 nM). Furthermore, two of the three mAbs (S2A3 and S2D5) bound only MERS-CoV (Erasmus Medical Center (EMC)) and not other CoVs, reflecting their high binding specificity. However, the mAbs lacked MERS-CoV neutralizing activity. Given their high affinity, specificity, and desirable stabilities, we anticipate that these anti-MERS-CoV mAbs would be suitable reagents for developing antibody-based diagnostics in laboratory or hospital settings for point-of-care testing.

18.
Mol Immunol ; 114: 62-71, 2019 10.
Article in English | MEDLINE | ID: mdl-31336250

ABSTRACT

The Fc region of IgG antibodies is crucial for binding to Fc receptors expressed on the surfaces of various immune leukocytes and eliciting therapeutic effector functions such as clearance of antibody-opsonized tumor cells. Despite abrogated Fc gamma receptor (FcγR) binding and therapeutic effector function in the absence of N-linked glycosylation at Asn297, the aglycosylated Fc region of IgG antibodies has bioprocessing advantages such as the absence of glycan heterogeneity and simple bacterial antibody production. Therefore, these antibodies have been comprehensively engineered as effector functional units for human therapy. In this work, we constructed a huge library of Fc variants with combinations of 25 beneficial mutations that were previously identified to improve binding of glycosylated or aglycosylated Fc regions to human FcγRs in previous studies. High-throughput screening of the resulting library led to the identification of an aglycosylated Fc variant that exhibited almost double the antibody-dependent cell-mediated cytotoxicity than wild-type glycosylated Fc. All mutations in this aglycosylated Fc variant were derived from previously identified beneficial mutations for engineered aglycosylated Fc variants as opposed to glycosylated variants, suggesting that significantly different sets of beneficial mutations are necessary to improve the effector function of aglycosylated Fc.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Mutation/immunology , Cell Line , Glycosylation , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Protein Binding/immunology , Protein Engineering , Receptors, IgG/immunology
19.
J Microbiol Biotechnol ; 29(5): 776-784, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31030455

ABSTRACT

Polyhydroxybutyrate (PHB), the most well-known polyhydroxyalkanoate, is a bio-based, biodegradable polymer that has the potential to replace petroleum-based plastics. Lignocellulose hydrolysate, a non-edible resource, is a promising substrate for the sustainable, fermentative production of PHB. However, its application is limited by the generation of inhibitors during the pretreatment processes. In this study, we investigated the feasibility of PHB production in E. coli in the presence of inhibitors found in lignocellulose hydrolysates. Our results show that the introduction of PHB synthetic genes (bktB, phaB, and phaC from Ralstonia eutropha H16) improved cell growth in the presence of the inhibitors such as furfural, 4-hydroxybenzaldehyde, and vanillin, suggesting that PHB synthetic genes confer resistance to these inhibitors. In addition, increased PHB production was observed in the presence of furfural as opposed to the absence of furfural, suggesting that this compound could be used to stimulate PHB production. Our findings indicate that PHB production using lignocellulose hydrolysates in recombinant E. coli could be an innovative strategy for cost-effective PHB production, and PHB could be a good target product from lignocellulose hydrolysates, especially glucose.


Subject(s)
Acclimatization/genetics , Escherichia coli/genetics , Furaldehyde/adverse effects , Genes, Synthetic/genetics , Hydroxybutyrates/metabolism , Polyesters/metabolism , Bacterial Proteins/genetics , Cupriavidus necator/genetics , Drug Resistance , Escherichia coli/growth & development , Escherichia coli/metabolism , Hordeum/enzymology , Lignin/metabolism , Pinus/enzymology , Poaceae/embryology
20.
Protein Expr Purif ; 159: 34-41, 2019 07.
Article in English | MEDLINE | ID: mdl-30880170

ABSTRACT

The plant protein production system is a platform that can not only reduce production costs but also produce monoclonal antibodies that do not have the risk of residual proteins from the host. However, due to the difference between post-translational processes in plants and animals, there may be a modification in the Fab region of the monoclonal antibody produced in the plant; thus, it is necessary to compare the antigen affinity of this antibody with that of the prototype. In this study, ofatumumab, a fully human anti-CD20 IgG1κ monoclonal antibody used for its non-cross resistance to rituximab, was expressed in Nicotiana benthamiana, and its affinities and efficacies were compared with those of native ofatumumab produced from CHO cells. Two forms of plant ofatumumab (with or without HDEL-tag) were generated and their production yields were compared. The HDEL-tagged ofatumumab was more expressed in plants than the form without HDEL-tag. The specificity of the target recognition of plant-derived ofatumumab was confirmed by mCherry-CD20-expressing HEK cells via immuno-staining, and the capping of CD20 after ofatumumab binding was also confirmed using Ramos B cells. In the functional equivalence tests, the binding affinities and complement-dependent cell cytotoxicity efficacy of plant-ofatumumab-HDEL and plant-ofatumumab without HDEL were significantly reduced compared to those of CHO-derived ofatumumab. Therefore, we suggest that although ofatumumab is not a good candidate as a template for plant-derived monoclonal antibodies because of its decreased affinity when produced in plants, it is an interesting target to study the differences between post-translational modifications in mammals and plants.


Subject(s)
Antibodies, Monoclonal, Humanized/genetics , Immunoglobulin Fab Fragments/chemistry , Nicotiana/metabolism , Plant Leaves/metabolism , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/metabolism , Antigens, CD20/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis , B-Lymphocytes , CHO Cells , Cell Death/drug effects , Cell Line, Tumor , Cricetulus , Cytotoxicity, Immunologic/drug effects , HEK293 Cells , Humans , Protein Conformation , Rituximab/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...