109 results on '"Inchan Kwon"'
Search Results
2. Enhanced therapeutic potential of antibody fragment via IEDDA-mediated site-specific albumin conjugation
- Author
-
Eun Byeol Go, Jae Hun Lee, Jeong Haeng Cho, Na Hyun Kwon, Jong-il Choi, and Inchan Kwon
- Subjects
Single-chain variable fragment ,Therapeutic antibody ,Human serum albumin ,Site-specific conjugation ,Half-life extension ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background The use of single-chain variable fragments (scFvs) for treating human diseases, such as cancer and immune system disorders, has attracted significant attention. However, a critical drawback of scFv is its extremely short serum half-life, which limits its therapeutic potential. Thus, there is a critical need to prolong the serum half-life of the scFv for clinical applications. One promising serum half-life extender for therapeutic proteins is human serum albumin (HSA), which is the most abundant protein in human serum, known to have an exceptionally long serum half-life. However, conjugating a macromolecular half-life extender to a small protein, such as scFv, often results in a significant loss of its critical properties. Results In this study, we conjugated the HSA to a permissive site of scFv to improve pharmacokinetic profiles. To ensure minimal damage to the antigen-binding capacity of scFv upon HSA conjugation, we employed a site-specific conjugation approach using a heterobifunctional crosslinker that facilitates thiol-maleimide reaction and inverse electron-demand Diels-Alder reaction (IEDDA). As a model protein, we selected 4D5scFv, derived from trastuzumab, a therapeutic antibody used in human epithermal growth factor 2 (HER2)-positive breast cancer treatment. We introduced a phenylalanine analog containing a very reactive tetrazine group (frTet) at conjugation site candidates predicted by computational methods. Using the linker TCO-PEG4-MAL, a single HSA molecule was site-specifically conjugated to the 4D5scFv (4D5scFv-HSA). The 4D5scFv-HSA conjugate exhibited HER2 binding affinity comparable to that of unmodified 4D5scFv. Furthermore, in pharmacokinetic profile in mice, the serum half-life of 4D5scFv-HSA was approximately 12 h, which is 85 times longer than that of 4D5scFv. Conclusions The antigen binding results and pharmacokinetic profile of 4D5scFv-HSA demonstrate that the site-specifically albumin-conjugated scFv retained its binding affinity with a prolonged serum half-life. In conclusion, we developed an effective strategy to prepare site-specifically albumin-conjugated 4D5scFv, which can have versatile clinical applications with improved efficacy.
- Published
- 2024
- Full Text
- View/download PDF
3. Recovery of rare earth elements from low-grade coal fly ash using a recyclable protein biosorbent
- Author
-
Zohaib Hussain, Divya Dwivedi, and Inchan Kwon
- Subjects
rare earth elements ,biosorption ,biomaterials ,elastin-like polypeptide ,liquid–liquid phase separation ,intrinsically disordered protein ,Biotechnology ,TP248.13-248.65 - Abstract
Rare earth elements (REEs), including those in the lanthanide series, are crucial components essential for clean energy transitions, but they originate from geographically limited regions. Exploiting new and diverse supply sources is vital to facilitating a clean energy future. Hence, we explored the recovery of REEs from coal fly ash (FA), a complex, low-grade industrial feedstock that is currently underutilized (leachate concentrations of REEs in FA are < 0.003 mol%). Herein, we demonstrated the thermo-responsive genetically encoded REE-selective elastin-like polypeptides (RELPs) as a recyclable bioengineered protein adsorbent for the selective retrieval of REEs from coal fly ash over multiple cycles. The results showed that RELPs could be efficiently separated using temperature cycling and reused with high stability, as they retained ∼95% of their initial REE binding capacity even after four cycles. Moreover, RELPs selectively recovered high-purity REEs from the simulated solution containing one representative REE in the range of 0.0001–0.005 mol%, resulting in up to a 100,000-fold increase in REE purity. This study offers a sustainable approach to diversifying REE supplies by recovering REEs from low-grade coal fly ash in industrial wastes and provides a scientific basis for the extraction of high-purity REEs for industrial purposes.
- Published
- 2024
- Full Text
- View/download PDF
4. In vivo study of newly developed albumin-conjugated urate oxidase for gout treatment
- Author
-
Jeonghaeng Cho, Byungseop Yang, Jae Hun Lee, Hyunwoo Kim, Hyeongseok Kim, Eun Byeol Go, Dong-ho Bak, Su Jin Park, Inchan Kwon, Jong-il Choi, and Kyunghee Lee
- Subjects
Gout ,Urate oxidase ,AfUox ,rHA ,Site-specific conjugation ,IEDDA ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Exogenously providing engineered Uox with enhanced half-life is one of the important urate-lowering treatments for gout. The potential of PAT101, a recombinant human albumin (rHA)-conjugated variant, was evaluated and compared as a novel gout treatment through various in vivo studies with PAT101 and competing drugs. Methods PAT101 was produced by site-specific conjugation of rHA and Aspergillus flavus Uox (AfUox-rHA) through clickable non-natural amino acid (frTet) and Inverse electron demand Diels–Alder (IEDDA) reaction. In vivo pharmacokinetics, efficacy tests and in vitro immunogenetic assay were performed after single or multiple doses of PAT101 and its competitors in BALB/c mice, transgenic (TG) mice, Sprague–Dawley (SD) rats, and non-human primate (NHP). Results The half-life of PAT101 in single-dose treated TG mice was more than doubled compared to pegloticase. In SD rats with 4 weeks of repeated administration of rasburicase, only 24% of Uox activity remained, whereas in PAT101, it was maintained by 86%. In the Uox KO model, the survival rate of PAT101 was comparable to that of pegloticase. In addition, human PBMC-based CD4+/CD8+ T-cell activation analysis demonstrated that PAT101 has a lower immune response compared to the original drug, rasburicase. Conclusion All results suggest that this rHA-conjugated AfUox, PAT101, can be provided as a reliable source of Uox for gout treatment.
- Published
- 2023
- Full Text
- View/download PDF
5. Real flue gas CO2 hydrogenation to formate by an enzymatic reactor using O2- and CO-tolerant hydrogenase and formate dehydrogenase
- Author
-
Jaehyun Cha, Jinhee Lee, Byoung Wook Jeon, Yong Hwan Kim, and Inchan Kwon
- Subjects
carbon dioxide ,hydrogen ,formate ,flue gas ,hydrogenase ,formate dehydrogenase ,Biotechnology ,TP248.13-248.65 - Abstract
It is challenging to capture carbon dioxide (CO2), a major greenhouse gas in the atmosphere, due to its high chemical stability. One potential practical solution to eliminate CO2 is to convert CO2 into formate using hydrogen (H2) (CO2 hydrogenation), which can be accomplished with inexpensive hydrogen from sustainable sources. While industrial flue gas could provide an adequate source of hydrogen, a suitable catalyst is needed that can tolerate other gas components, such as carbon monoxide (CO) and oxygen (O2), potential inhibitors. Our proposed CO2 hydrogenation system uses the hydrogenase derived from Ralstonia eutropha H16 (ReSH) and formate dehydrogenase derived from Methylobacterium extorquens AM1 (MeFDH1). Both enzymes are tolerant to CO and O2, which are typical inhibitors of metalloenzymes found in flue gas. We have successfully demonstrated that combining ReSH- and MeFDH1-immobilized resins can convert H2 and CO2 in real flue gas to formate via a nicotinamide adenine dinucleotide-dependent cascade reaction. We anticipated that this enzyme system would enable the utilization of diverse H2 and CO2 sources, including waste gases, biomass, and gasified plastics.
- Published
- 2023
- Full Text
- View/download PDF
6. Hydrogen-fueled CO2 reduction using oxygen-tolerant oxidoreductases
- Author
-
Jaehyun Cha, Hyeonseon Bak, and Inchan Kwon
- Subjects
hydrogen ,carbon dioxide ,formate ,oxygen-tolerant ,hydrogenase ,formate dehydrogenase ,Biotechnology ,TP248.13-248.65 - Abstract
Hydrogen gas obtained from cheap or sustainable sources has been investigated as an alternative to fossil fuels. By using hydrogenase (H2ase) and formate dehydrogenase (FDH), H2 and CO2 gases can be converted to formate, which can be conveniently stored and transported. However, developing an enzymatic process that converts H2 and CO2 obtained from cheap sources into formate is challenging because even a very small amount of O2 included in the cheap sources damages most H2ases and FDHs. In order to overcome this limitation, we investigated a pair of oxygen-tolerant H2ase and FDH. We achieved the cascade reaction between H2ase from Ralstonia eutropha H16 (ReSH) and FDH from Rhodobacter capsulatus (RcFDH) to convert H2 and CO2 to formate using in situ regeneration of NAD+/NADH in the presence of O2.
- Published
- 2023
- Full Text
- View/download PDF
7. Computation-Aided Design of Albumin Affibody-Inserted Antibody Fragment for the Prolonged Serum Half-Life
- Author
-
Na Hyun Kwon, Jae Hun Lee, and Inchan Kwon
- Subjects
single-chain antibody fragment ,albumin affibody ,half-life extension ,fusion protein ,structure prediction ,cancer ,Pharmacy and materia medica ,RS1-441 - Abstract
Single-chain variable fragments (scFvs) have been recognized as promising agents in cancer therapy. However, short serum half-life of scFvs often limits clinical application. Fusion to albumin affibody (ABD) is an effective and convenient half-life extension strategy. Although one terminus of scFv is available for fusion of ABD, it is also frequently used for fusion of useful moieties such as small functional proteins, cytokines, or antibodies. Herein, we investigated the internal linker region for ABD fusion instead of terminal region, which was rarely explored before. We constructed two internally ABD-inserted anti-HER2 4D5scFv (4D5-ABD) variants, which have short (4D5-S-ABD) and long (4D5-L-ABD) linker length respectively. The model structures of these 4D5scFv and 4D5-ABD variants predicted using the deep learning-based protein structure prediction program (AlphaFold2) revealed high similarity to either the original 4D5scFv or the ABD structure, implying that the functionality would be retained. Designed 4D5-ABD variants were expressed in the bacterial expression system and characterized. Both 4D5-ABD variants showed anti-HER2 binding affinity comparable with 4D5scFv. Binding affinity of both 4D5-ABD variants against albumin was also comparable. In a pharmacokinetic study in mice, the 4D5-ABD variants showed a significantly prolonged half-life of 34 h, 114 times longer than that of 4D5scFv. In conclusion, we have developed a versatile scFv platform with enhanced pharmacokinetic profiles with an aid of deep learning-based structure prediction.
- Published
- 2022
- Full Text
- View/download PDF
8. Chemical Modification of Cysteine with 3-Arylpropriolonitrile Improves the In Vivo Stability of Albumin-Conjugated Urate Oxidase Therapeutic Protein
- Author
-
Byungseop Yang and Inchan Kwon
- Subjects
thiol-maleimide ,3-arylpropiolonitriles ,half-life extension ,site-specific albumin conjugation ,urate oxidase ,therapeutic protein ,Biology (General) ,QH301-705.5 - Abstract
3-arylpropiolonitriles (APN) are promising alternatives to maleimide for chemo-selective thiol conjugation, because the reaction product has a remarkably hydrolytic stability compared with that of thiol-maleimide reactions in vitro. However, whether cysteine modification with APN enhances stability in vivo compared to thiol-maleimide reactions remains unclear, probably due to the too short in vivo serum half-life of a protein to observe significant cleavage of thiol-maleimide/-APN reaction products. The conjugation of human serum albumin (HSA) to a therapeutic protein reportedly prolongs the in vivo serum half-life. To evaluate the in vivo stability of the thiol-APN reaction product, we prepared HSA-conjugated Arthrobacter globiformis urate oxidase (AgUox), a therapeutic protein for gout treatment. Site-specific HSA conjugation to AgUox was achieved by combining site-specific incorporation of tetrazine containing an amino acid (frTet) into AgUox and a crosslinker containing trans-cyclooctene and either thiol-maleimide (AgUox-MAL-HSA) or -APN chemistry (AgUox-APN-HSA). Substantial cleavage of the thioester of AgUox-MAL-HSA was observed in vitro, whereas no cleavage of the thiol-APN product of AgUox-APN-HSA was observed. Furthermore, the in vivo serum half-life of AgUox-APN-HSA in the late phase was significantly longer than that of AgUox-MAL-HSA. Overall, these results demonstrate that the thiol-APN chemistry enhanced the in vivo stability of the HSA-conjugated therapeutic protein.
- Published
- 2021
- Full Text
- View/download PDF
9. Thermostable and Long-Circulating Albumin-Conjugated Arthrobacter globiformis Urate Oxidase
- Author
-
Byungseop Yang and Inchan Kwon
- Subjects
Arthrobacter globiformis ,gout ,half-life extension ,inverse electron demand Diels-Alder reaction ,site-specific albumin conjugation ,thermostability ,Pharmacy and materia medica ,RS1-441 - Abstract
Urate oxidase derived from Aspergillus flavus has been investigated as a treatment for tumor lysis syndrome, hyperuricemia, and gout. However, its long-term use is limited owing to potential immunogenicity, low thermostability, and short circulation time in vivo. Recently, urate oxidase isolated from Arthrobacter globiformis (AgUox) has been reported to be thermostable and less immunogenic than the Aspergillus-derived urate oxidase. Conjugation of human serum albumin (HSA) to therapeutic proteins has become a promising strategy to prolong circulation time in vivo. To develop a thermostable and long-circulating urate oxidase, we investigated the site-specific conjugation of HSA to AgUox based on site-specific incorporation of a clickable non-natural amino acid (frTet) and an inverse electron demand Diels–Alder reaction. We selected 14 sites for frTet incorporation using the ROSETTA design, a computational stability prediction program, among which AgUox containing frTet at position 196 (Ag12) exhibited enzymatic activity and thermostability comparable to those of wild-type AgUox. Furthermore, Ag12 exhibited a high HSA conjugation yield without compromising the enzymatic activity, generating well-defined HSA-conjugated AgUox (Ag12-HSA). In mice, the serum half-life of Ag12-HSA was approximately 29 h, which was roughly 17-fold longer than that of wild-type AgUox. Altogether, this novel formulated AgUox may hold enhanced therapeutic efficacy for several diseases.
- Published
- 2021
- Full Text
- View/download PDF
10. Effect of C-terminus Conjugation via Different Conjugation Chemistries on In Vivo Activity of Albumin-Conjugated Recombinant GLP-1
- Author
-
Junyong Park, Mijeong Bak, Kiyoon Min, Hyun-Woo Kim, Jeong-Haeng Cho, Giyoong Tae, and Inchan Kwon
- Subjects
plasma half-life extension ,albumin conjugation ,in vivo glucose-lowering activity ,glucagon-like peptide-1 ,Pharmacy and materia medica ,RS1-441 - Abstract
Glucagon-like peptide-1 (GLP-1) is a peptide hormone with tremendous therapeutic potential for treating type 2 diabetes mellitus. However, the short half-life of its native form is a significant drawback. We previously prolonged the plasma half-life of GLP-1 via site-specific conjugation of human serum albumin (HSA) at position 16 of recombinant GLP-1 using site-specific incorporation of p-azido-phenylalanine (AzF) and strain-promoted azide-alkyne cycloaddition (SPAAC). However, the resulting conjugate GLP1_8G16AzF-HSA showed only moderate in vivo glucose-lowering activity, probably due to perturbed interactions with GLP-1 receptor (GLP-1R) caused by the albumin-linker. To identify albumin-conjugated GLP-1 variants with enhanced in vivo glucose-lowering activity, we investigated the conjugation of HSA to a C-terminal region of GLP-1 to reduce steric hindrance by the albumin-linker using two different conjugation chemistries. GLP-1 variants GLP1_8G37AzF-HSA and GLP1_8G37C-HSA were prepared using SPAAC and Michael addition, respectively. GLP1_8G37C-HSA exhibited a higher glucose-lowering activity in vivo than GLP1_8G16AzF-HSA, while GLP1_8G37AzF-HSA did not. Another GLP-1 variant, GLP1_8A37C-HSA, had a glycine to alanine mutation at position 8 and albumin at its C-terminus and exhibited in vivo glucose-lowering activity comparable to that of GLP1_8G37C-HSA, despite a moderately shorter plasma half-life. These results showed that site-specific HSA conjugation to the C-terminus of GLP-1 via Michael addition could be used to generate GLP-1 variants with enhanced glucose-lowering activity and prolonged plasma half-life in vivo.
- Published
- 2021
- Full Text
- View/download PDF
11. Recombinant Peptide Production Platform Coupled with Site-Specific Albumin Conjugation Enables a Convenient Production of Long-Acting Therapeutic Peptide
- Author
-
Mijeong Bak, Junyong Park, Kiyoon Min, Jinhwan Cho, Jihyoun Seong, Young S. Hahn, Giyoong Tae, and Inchan Kwon
- Subjects
recombinant therapeutic peptide ,serum half-life extension ,albumin conjugation ,Pharmacy and materia medica ,RS1-441 - Abstract
The number of therapeutic peptides for human treatment is growing rapidly. However, their development faces two major issues: the poor yield of large peptides from conventional solid-phase synthesis, and the intrinsically short serum half-life of peptides. To address these issues, we investigated a platform for the production of a recombinant therapeutic peptide with an extended serum half-life involving the site-specific conjugation of human serum albumin (HSA). HSA has an exceptionally long serum half-life and can be used to extend the serum half-lives of therapeutic proteins and peptides. We used glucagon-like-peptide 1 (GLP-1) as a model peptide in the present study. A “clickable” non-natural amino acid—p-azido-l-phenylalanine (AzF)—was incorporated into three specific sites (V16, Y19, and F28) of a GLP-1 variant, followed by conjugation with HSA through strain-promoted azide–alkyne cycloaddition. All three HSA-conjugated GLP-1 variants (GLP1_16HSA, GLP1_19HSA, and GLP1_28HSA) exhibited comparable serum half-lives in vivo. However, the three GLP1_HSA variants had different in vitro biological activities and in vivo glucose-lowering effects, demonstrating the importance of site-specific HSA conjugation. The platform described herein could be used to develop other therapeutic peptides with extended serum half-lives.
- Published
- 2020
- Full Text
- View/download PDF
12. The Minimal Effect of Linker Length for Fatty Acid Conjugation to a Small Protein on the Serum Half-Life Extension
- Author
-
Jinhwan Cho, Junyong Park, Giyoong Tae, Mi Sun Jin, and Inchan Kwon
- Subjects
serum half-life extension ,fatty acid conjugation ,FcRn-mediated recycling ,serum albumin ,Biology (General) ,QH301-705.5 - Abstract
Conjugation of serum albumin or one of its ligands (such as fatty acid) has been an effective strategy to prolong the serum half-lives of drugs via neonatal Fc receptor (FcRn)–mediated recycling of albumin. So far, fatty acid (FA) has been effective in prolonging the serum half-lives for therapeutic peptides and small proteins, but not for large therapeutic proteins. Very recently, it was reported a large protein conjugated to FA competes with the binding of FcRn with serum albumin, leading to limited serum half-life extension, because primary FA binding sites in serum albumin partially overlap with FcRn binding sites. In order to prevent such competition, longer linkers between FA and the large proteins were required. Herein, we hypothesized that small proteins do not cause substantial competition for FcRn binding to albumin, resulting in the extended serum half-life. Using a small protein (28 kDa), we investigated whether the intramolecular distance in FA-protein conjugate affects the FcRn binding with albumin and serum half-life using linkers with varying lengths. Unlike with the FA-conjugated large protein, all FA-conjugated small proteins with different linkers exhibited comparable the FcRn binding to albumin and extended serum half-life.
- Published
- 2020
- Full Text
- View/download PDF
13. Site-Specific Bioconjugation of an Organometallic Electron Mediator to an Enzyme with Retained Photocatalytic Cofactor Regenerating Capacity and Enzymatic Activity
- Author
-
Sung In Lim, Sungho Yoon, Yong Hwan Kim, and Inchan Kwon
- Subjects
electron mediator ,non-natural amino acid ,redox enzyme ,site-specific bioconjugation ,formate dehydrogenase ,Organic chemistry ,QD241-441 - Abstract
Photosynthesis consists of a series of reactions catalyzed by redox enzymes to synthesize carbohydrates using solar energy. In order to take the advantage of solar energy, many researchers have investigated artificial photosynthesis systems mimicking the natural photosynthetic enzymatic redox reactions. These redox reactions usually require cofactors, which due to their high cost become a key issue when constructing an artificial photosynthesis system. Combining a photosensitizer and an Rh-based electron mediator (RhM) has been shown to photocatalytically regenerate cofactors. However, maintaining the high concentration of cofactors available for efficient enzymatic reactions requires a high concentration of the expensive RhM; making this process cost prohibitive. We hypothesized that conjugation of an electron mediator to a redox enzyme will reduce the amount of electron mediators necessary for efficient enzymatic reactions. This is due to photocatalytically regenerated NAD(P)H being readily available to a redox enzyme, when the local NAD(P)H concentration near the enzyme becomes higher. However, conventional random conjugation of RhM to a redox enzyme will likely lead to a substantial loss of cofactor regenerating capacity and enzymatic activity. In order to avoid this issue, we investigated whether bioconjugation of RhM to a permissive site of a redox enzyme retains cofactor regenerating capacity and enzymatic activity. As a model system, a RhM was conjugated to a redox enzyme, formate dehydrogenase obtained from Thiobacillus sp. KNK65MA (TsFDH). A RhM-containing azide group was site-specifically conjugated to p-azidophenylalanine introduced to a permissive site of TsFDH via a bioorthogonal strain-promoted azide-alkyne cycloaddition and an appropriate linker. The TsFDH-RhM conjugate exhibited retained cofactor regenerating capacity and enzymatic activity.
- Published
- 2015
- Full Text
- View/download PDF
14. Forced Ambiguity of the Leucine Codons for Multiple-Site-Specific Incorporation of a Noncanonical Amino Acid.
- Author
-
Inchan Kwon and Eun Sil Choi
- Subjects
Medicine ,Science - Abstract
Multiple-site-specific incorporation of a noncanonical amino acid into a recombinant protein would be a very useful technique to generate multiple chemical handles for bioconjugation and multivalent binding sites for the enhanced interaction. Previously combination of a mutant yeast phenylalanyl-tRNA synthetase variant and the yeast phenylalanyl-tRNA containing the AAA anticodon was used to incorporate a noncanonical amino acid into multiple UUU phenylalanine (Phe) codons in a site-specific manner. However, due to the less selective codon recognition of the AAA anticodon, there was significant misincorporation of a noncanonical amino acid into unwanted UUC Phe codons. To enhance codon selectivity, we explored degenerate leucine (Leu) codons instead of Phe degenerate codons. Combined use of the mutant yeast phenylalanyl-tRNA containing the CAA anticodon and the yPheRS_naph variant allowed incorporation of a phenylalanine analog, 2-naphthylalanine, into murine dihydrofolate reductase in response to multiple UUG Leu codons, but not to other Leu codon sites. Despite the moderate UUG codon occupancy by 2-naphthylalaine, these results successfully demonstrated that the concept of forced ambiguity of the genetic code can be achieved for the Leu codons, available for multiple-site-specific incorporation.
- Published
- 2016
- Full Text
- View/download PDF
15. Site-specific bioconjugation of a murine dihydrofolate reductase enzyme by copper(I)-catalyzed azide-alkyne cycloaddition with retained activity.
- Author
-
Sung In Lim, Yukina Mizuta, Akinori Takasu, Yong Hwan Kim, and Inchan Kwon
- Subjects
Medicine ,Science - Abstract
Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) is an efficient reaction linking an azido and an alkynyl group in the presence of copper catalyst. Incorporation of a non-natural amino acid (NAA) containing either an azido or an alkynyl group into a protein allows site-specific bioconjugation in mild conditions via CuAAC. Despite its great potential, bioconjugation of an enzyme has been hampered by several issues including low yield, poor solubility of a ligand, and protein structural/functional perturbation by CuAAC components. In the present study, we incorporated an alkyne-bearing NAA into an enzyme, murine dihydrofolate reductase (mDHFR), in high cell density cultivation of Escherichia coli, and performed CuAAC conjugation with fluorescent azide dyes to evaluate enzyme compatibility of various CuAAC conditions comprising combination of commercially available Cu(I)-chelating ligands and reductants. The condensed culture improves the protein yield 19-fold based on the same amount of non-natural amino acid, and the enzyme incubation under the optimized reaction condition did not lead to any activity loss but allowed a fast and high-yield bioconjugation. Using the established conditions, a biotin-azide spacer was efficiently conjugated to mDHFR with retained activity leading to the site-specific immobilization of the biotin-conjugated mDHFR on a streptavidin-coated plate. These results demonstrate that the combination of reactive non-natural amino acid incorporation and the optimized CuAAC can be used to bioconjugate enzymes with retained enzymatic activity.
- Published
- 2014
- Full Text
- View/download PDF
16. Halogenation generates effective modulators of amyloid-Beta aggregation and neurotoxicity.
- Author
-
H Edward Wong, Jacob A Irwin, and Inchan Kwon
- Subjects
Medicine ,Science - Abstract
Halogenation of organic compounds plays diverse roles in biochemistry, including selective chemical modification of proteins and improved oral absorption/blood-brain barrier permeability of drug candidates. Moreover, halogenation of aromatic molecules greatly affects aromatic interaction-mediated self-assembly processes, including amyloid fibril formation. Perturbation of the aromatic interaction caused by halogenation of peptide building blocks is known to affect the morphology and other physical properties of the fibrillar structure. Consequently, in this article, we investigated the ability of halogenated ligands to modulate the self-assembly of amyloidogenic peptide/protein. As a model system, we chose amyloid-beta peptide (Aβ), which is implicated in Alzheimer's disease, and a novel modulator of Aβ aggregation, erythrosine B (ERB). Considering that four halogen atoms are attached to the xanthene benzoate group in ERB, we hypothesized that halogenation of the xanthene benzoate plays a critical role in modulating Aβ aggregation and cytotoxicity. Therefore, we evaluated the modulating capacities of four ERB analogs containing different types and numbers of halogen atoms as well as fluorescein as a negative control. We found that fluorescein is not an effective modulator of Aβ aggregation and cytotoxicity. However, halogenation of either the xanthenes or benzoate ring of fluorescein substantially enhanced the inhibitory capacity on Aβ aggregation. Such Aβ aggregation inhibition by ERB analogs except rose bengal correlated well to the inhibition of Aβ cytotoxicity. To our knowledge, this is the first report demonstrating that halogenation of aromatic rings substantially enhance inhibitory capacities of small molecules on Aβ-associated neurotoxicity via Aβ aggregation modulation.
- Published
- 2013
- Full Text
- View/download PDF
17. Xanthene food dye, as a modulator of Alzheimer's disease amyloid-beta peptide aggregation and the associated impaired neuronal cell function.
- Author
-
H Edward Wong and Inchan Kwon
- Subjects
Medicine ,Science - Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD is a degenerative brain disorder that causes problems with memory, thinking and behavior. It has been suggested that aggregation of amyloid-beta peptide (Aβ) is closely linked to the development of AD pathology. In the search for safe, effective modulators, we evaluated the modulating capabilities of erythrosine B (ER), a Food and Drug Administration (FDA)-approved red food dye, on Aβ aggregation and Aβ-associated impaired neuronal cell function.In order to evaluate the modulating ability of ER on Aβ aggregation, we employed transmission electron microscopy (TEM), thioflavin T (ThT) fluorescence assay, and immunoassays using Aβ-specific antibodies. TEM images and ThT fluorescence of Aβ samples indicate that protofibrils are predominantly generated and persist for at least 3 days. The average length of the ER-induced protofibrils is inversely proportional to the concentration of ER above the stoichiometric concentration of Aβ monomers. Immunoassay results using Aβ-specific antibodies suggest that ER binds to the N-terminus of Aβ and inhibits amyloid fibril formation. In order to evaluate Aβ-associated toxicity we determined the reducing activity of SH-SY5Y neuroblastoma cells treated with Aβ aggregates formed in the absence or in the presence of ER. As the concentration of ER increased above the stoichiometric concentration of Aβ, cellular reducing activity increased and Aβ-associated reducing activity loss was negligible at 500 µM ER.Our findings show that ER is a novel modulator of Aβ aggregation and reduces Aβ-associated impaired cell function. Our findings also suggest that xanthene dye can be a new type of small molecule modulator of Aβ aggregation. With demonstrated safety profiles and blood-brain permeability, ER represents a particularly attractive aggregation modulator for amyloidogenic proteins associated with neurodegenerative diseases.
- Published
- 2011
- Full Text
- View/download PDF
18. Recovery of rare earth elements from low-grade coal fly ash using a recyclable protein biosorbent.
- Author
-
Hussain, Zohaib, Dwivedi, Divya, and Inchan Kwon
- Published
- 2024
- Full Text
- View/download PDF
19. Self-Sufficient Reusable Biocatalytic System Outfitted with Multiple Oxidoreductases and Flexible Polypeptide-Based Cofactor Swing Arms
- Author
-
Jaehyun Cha, Jinhwan Cho, and Inchan Kwon
- Subjects
Renewable Energy, Sustainability and the Environment ,General Chemical Engineering ,Environmental Chemistry ,General Chemistry - Published
- 2023
- Full Text
- View/download PDF
20. Real Flue Gas Co2 Hydrogenation to Formate by an Enzymatic Reactor Using Rare O2- and Co-Tolerant Hydrogenase and Formate Dehydrogenase
- Author
-
Jaehyun Cha, Jinhee Lee, Byoung Wook Jeon, Yong Hwan Kim, and Inchan Kwon
- Published
- 2023
- Full Text
- View/download PDF
21. Multivalent Albumin–Neonatal Fc Receptor Interactions Mediate a Prominent Extension of the Serum Half-Life of a Therapeutic Protein
- Author
-
Byungseop Yang and Inchan Kwon
- Subjects
Urate Oxidase ,Protein subunit ,Serum albumin ,Pharmaceutical Science ,Serum Albumin, Human ,Receptors, Fc ,02 engineering and technology ,Conjugated system ,030226 pharmacology & pharmacy ,Excipients ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neonatal Fc receptor ,Drug Discovery ,medicine ,Animals ,Enzyme Assays ,Cycloaddition Reaction ,biology ,Chemistry ,Histocompatibility Antigens Class I ,Albumin ,021001 nanoscience & nanotechnology ,Human serum albumin ,body regions ,Injections, Intravenous ,embryonic structures ,biology.protein ,Biophysics ,Molecular Medicine ,Female ,0210 nano-technology ,Half-Life ,medicine.drug ,Phenylalanine analog ,Conjugate - Abstract
Human serum albumin (HSA) has been used to extend the serum half-life of therapeutic proteins owing to its exceptionally long serum half-life via the neonatal Fc receptor (FcRn)-mediated recycling mechanism. In most cases, only one HSA molecule was conjugated to a therapeutic protein, leading to a limited extension of the serum half-life. In this study, we hypothesized that conjugation of multiple HSA molecules to a therapeutic protein significantly further extends the serum half-life via multivalent HSA-FcRn interactions. We chose urate oxidase (Uox), a tetrameric therapeutic protein used for the treatment of gout, as a model. In previous studies, only one HSA molecule was site-specifically conjugated to one Uox because of poor conjugation yield of the relatively slow bio-orthogonal chemistry, strain-promoted azide-alkyne cycloaddition (SPAAC). To increase the number of HSA molecules conjugated to one Uox, we employed the faster bio-orthogonal chemistry, inverse electron demand Diels-Alder reaction (IEDDA). We site-specifically introduced the phenylalanine analog with a fast-reacting tetrazine group (frTet) into position 174 of each subunit of Uox. We then achieved site-specific HSA conjugation to each subunit of Uox via IEDDA, generating Uox conjugated to four HSA molecules (Uox-HSA4), with a small portion of Uox conjugated to three HSA molecules (Uox-HSA3). We characterized Uox-HSA4 as well as Uox variants conjugated to one or two HSA molecules prepared via SPAAC (Uox-HSA1 or Uox-HSA2). The enzyme activity of all three Uox-HSA conjugates was comparable to that of unmodified Uox. We found out that an increase in HSA molecules conjugated to Uox (multiple albumin-conjugated therapeutic protein) enhanced FcRn binding and consequently prolonged the serum half-life in vivo. In particular, the conjugation of four HSA molecules to Uox led to a prominent extension of the serum half-life (over 21 h), which is about 16-fold longer than that of Uox-WT.
- Published
- 2021
- Full Text
- View/download PDF
22. Nano-Entrapping Multiple Oxidoreductases and Cofactor for All-In-One Nanoreactors
- Author
-
Inchan Kwon, Seoungkyun Kim, Kiyoon Kwon, and Giyoong Tae
- Subjects
chemistry.chemical_classification ,biology ,Renewable Energy, Sustainability and the Environment ,General Chemical Engineering ,02 engineering and technology ,General Chemistry ,Nanoreactor ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Combinatorial chemistry ,Redox ,Cofactor ,0104 chemical sciences ,Enzyme ,chemistry ,Oxidoreductase ,Nano ,biology.protein ,Environmental Chemistry ,0210 nano-technology - Abstract
Oxidoreductases have been used for the synthesis of value-added chemicals via redox reactions under mild conditions. However, these enzymes often require cofactors, which are expensive and small mo...
- Published
- 2021
- Full Text
- View/download PDF
23. Temporal Control of Efficient In Vivo Bioconjugation Using a Genetically Encoded Tetrazine-Mediated Inverse-Electron-Demand Diels–Alder Reaction
- Author
-
Byungseop Yang, Seoungkyun Kim, Ryan A. Mehl, Kiyoon Kwon, Giyoong Tae, Subhashis Jana, Inchan Kwon, and Savanna Avila-Crump
- Subjects
Pharmacology ,Bioconjugation ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,02 engineering and technology ,021001 nanoscience & nanotechnology ,01 natural sciences ,Fluorescence ,Coupling reaction ,0104 chemical sciences ,Green fluorescent protein ,Tetrazine ,chemistry.chemical_compound ,In vivo ,Biophysics ,Nanocarriers ,0210 nano-technology ,Inverse electron-demand Diels–Alder reaction ,Biotechnology - Abstract
An inverse-electron-demand Diels-Alder (IEDDA) reaction using genetically encoded tetrazine variants enables rapid bioconjugation for diverse applications in vitro and in cellulo. However, in vivo bioconjugation using genetically encoded tetrazine variants is challenging, because the IEDDA coupling reaction competes with rapid elimination of reaction partners in vivo. Here, we tested the hypothesis that a genetically encoded phenylalanine analogue containing a hydrogen-substituted tetrazine (frTet) would increase the IEDDA reaction rate, thereby allowing for successful bioconjugation in vivo. We found that the in vitro IEDDA reaction rate of superfolder green fluorescent protein (sfGFP) containing frTet (sfGFP-frTet) was 12-fold greater than that of sfGFP containing methyl-substituted tetrazine (sfGFP-Tet_v2.0). Additionally, sfGFP variants encapsulated with chitosan-modified, pluronic-based nanocarriers were delivered into nude mice or tumor-bearing mice for in vivo imaging. The in vivo-delivered sfGFP-frTet exhibited almost complete fluorescence recovery upon addition of trans-cyclooctene via the IEDDA reaction within 2 h, whereas sfGFP-Tet_v2.0 did not show substantial fluorescence recovery. These results demonstrated that the genetically encoded frTet allows an almost complete IEDDA reaction in vivo upon addition of trans-cyclooctene, enabling temporal control of in vivo bioconjugation in a very high yield.
- Published
- 2020
- Full Text
- View/download PDF
24. Pluronic-Based Nanocarrier Platform Encapsulating Two Enzymes for Cascade Reactions
- Author
-
Seoungkyun Kim, Jaehyun Cha, Kiyoon Kwon, Min Su Han, Soyeon Yoo, Inchan Kwon, and Giyoong Tae
- Subjects
chemistry.chemical_classification ,Immobilized enzyme ,Biochemistry (medical) ,Biomedical Engineering ,General Chemistry ,respiratory system ,Poloxamer ,Formate dehydrogenase ,Biomaterials ,Enzyme ,chemistry ,Mannitol dehydrogenase ,Biochemistry ,Cascade reaction ,Nanocarriers ,human activities ,Biosensor - Abstract
Enzyme immobilization is very important for diverse enzyme applications. Particularly, there is a growing need for coimmobilization of multiple enzymes for biosensing and synthetic applications. However, it is still challenging to coimmobilize two enzymes with desirable features, including high immobilization yield, retention of enzymatic activity, and low leaching. In this study, we demonstrated that a pluronic-based nanocarrier (PNC) can be an encapsulation platform for immobilization of various single enzymes. Since the PNC is temperature-sensitive, a simple temperature change from 4 to 37 °C led to a substantial size reduction and enzyme encapsulation. All six enzymes tested were encapsulated by the PNC in high yield (∼90%) with the retained enzymatic activity (95%). The leaching of encapsulated enzymes was very minimal (0.13% for 2 weeks). Then, we demonstrated that the PNC can efficiently coencapsulate two enzymes, formate dehydrogenase (FDH) and mannitol dehydrogenase (MDH), for a cascade reaction producing d-mannitol. Coencapsulation of FDH and MDH resulted in an over 10-fold increase in d-mannitol production compared to the free mix of FDH and MDH, likely due to the enhanced local concentrations of FDH and MDH inside the PNC.
- Published
- 2020
- Full Text
- View/download PDF
25. Intramolecular distance in the conjugate of urate oxidase and fatty acid governs FcRn binding and serum half-life in vivo
- Author
-
Jong Chul Kim, Giyoong Tae, Junyong Park, Inchan Kwon, Mi Sun Jin, Songwon Kim, and Jinhwan Cho
- Subjects
Urate Oxidase ,Serum albumin ,Pharmaceutical Science ,Hyperuricemia ,Receptors, Fc ,02 engineering and technology ,Mice ,03 medical and health sciences ,Neonatal Fc receptor ,In vivo ,Animals ,Serum Albumin ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Fatty Acids ,Histocompatibility Antigens Class I ,Urate oxidase ,021001 nanoscience & nanotechnology ,Ligand (biochemistry) ,Protein tertiary structure ,Biochemistry ,biology.protein ,0210 nano-technology ,Linker ,Half-Life ,Conjugate - Abstract
Therapeutic proteins are indispensable for treatment of various human diseases. However, intrinsic short serum half-lives of proteins are still big hurdles for developing new therapeutic proteins or expanding applications of existing ones. Urate oxidase (Uox) is a therapeutic protein clinically used for treatment of hyperuricemia. Due to its short half-life, its application for gout treatment requires prolonging the half-life in vivo. Conjugation of a fatty acid (FA), a serum albumin (SA) ligand, to therapeutic proteins/peptides is an emerging strategy to prolong serum half-life presumably via neonatal Fc receptor (FcRn)–mediated recycling. FA conjugation was proven effective for peptides and small proteins (less than 28 kDa), but not for Uox (140 kDa). We hypothesized that the intramolecular distance in the conjugate of FA and Uox is a critical factor for effective FcRn-mediated recycling. In order to control the intramolecular distance in the conjugate, we varied linker lengths between Uox and palmitic acid (PA). There was a linear correlation between the linker length and serum half-life of PA–conjugated Uox (Uox-PA) conjugates. The longer linker led to about 7-fold greater extension of serum half-life of Uox in mice than the unmodified Uox. The trend in serum half-life extension matched well with that in the tertiary structure formation of FcRn/SA/Uox-PA in vitro. These results demonstrate that the intramolecular distance in the conjugate of Uox and FA governs the stable formation of FcRn/SA/FA-conjugated protein and serum half-life extension in vivo. These findings would also contribute to development of effective FAconjugated therapeutic proteins.
- Published
- 2020
- Full Text
- View/download PDF
26. Charge Booster Tags for Controlled Release of Therapeutics from a Therapeutic Carrier (Adv. Funct. Mater. 11/2023)
- Author
-
Seoungkyun Kim, Dong Hee Kim, Jinhwan Cho, Jaeyun Kim, and Inchan Kwon
- Subjects
Biomaterials ,Electrochemistry ,Condensed Matter Physics ,Electronic, Optical and Magnetic Materials - Published
- 2023
- Full Text
- View/download PDF
27. Self-Sufficient Reusable Microreactor Outfitted with Multiple Oxidoreductases and Flexible Polypeptide-Based Cofactor Swing Arms
- Author
-
Jaehyun Cha, Jinhwan Cho, and Inchan Kwon
- Published
- 2022
- Full Text
- View/download PDF
28. Charge Booster Tags for Controlled Release of Therapeutics from a Therapeutic Carrier
- Author
-
Seoungkyun Kim, Dong Hee Kim, Jinhwan Cho, Jaeyun Kim, and Inchan Kwon
- Subjects
Biomaterials ,Electrochemistry ,Condensed Matter Physics ,Electronic, Optical and Magnetic Materials - Published
- 2022
- Full Text
- View/download PDF
29. Effect of C-terminus Conjugation via Different Conjugation Chemistries on In Vivo Activity of Albumin-Conjugated Recombinant GLP-1
- Author
-
Kiyoon Min, Hyun-Woo Kim, Inchan Kwon, Junyong Park, Giyoong Tae, Jeong-Haeng Cho, and Mijeong Bak
- Subjects
endocrine system ,Pharmaceutical Science ,lcsh:RS1-441 ,02 engineering and technology ,in vivo glucose-lowering activity ,Article ,albumin conjugation ,law.invention ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,In vivo ,law ,medicine ,Receptor ,030304 developmental biology ,Alanine ,0303 health sciences ,Chemistry ,digestive, oral, and skin physiology ,Albumin ,plasma half-life extension ,021001 nanoscience & nanotechnology ,Human serum albumin ,body regions ,Biochemistry ,glucagon-like peptide-1 ,Glycine ,embryonic structures ,Recombinant DNA ,0210 nano-technology ,medicine.drug ,Conjugate - Abstract
Glucagon-like peptide-1 (GLP-1) is a peptide hormone with tremendous therapeutic potential for treating type 2 diabetes mellitus. However, the short half-life of its native form is a significant drawback. We previously prolonged the plasma half-life of GLP-1 via site-specific conjugation of human serum albumin (HSA) at position 16 of recombinant GLP-1 using site-specific incorporation of p-azido-phenylalanine (AzF) and strain-promoted azide-alkyne cycloaddition (SPAAC). However, the resulting conjugate GLP1_8G16AzF-HSA showed only moderate in vivo glucose-lowering activity, probably due to perturbed interactions with GLP-1 receptor (GLP-1R) caused by the albumin-linker. To identify albumin-conjugated GLP-1 variants with enhanced in vivo glucose-lowering activity, we investigated the conjugation of HSA to a C-terminal region of GLP-1 to reduce steric hindrance by the albumin-linker using two different conjugation chemistries. GLP-1 variants GLP1_8G37AzF-HSA and GLP1_8G37C-HSA were prepared using SPAAC and Michael addition, respectively. GLP1_8G37C-HSA exhibited a higher glucose-lowering activity in vivo than GLP1_8G16AzF-HSA, while GLP1_8G37AzF-HSA did not. Another GLP-1 variant, GLP1_8A37C-HSA, had a glycine to alanine mutation at position 8 and albumin at its C-terminus and exhibited in vivo glucose-lowering activity comparable to that of GLP1_8G37C-HSA, despite a moderately shorter plasma half-life. These results showed that site-specific HSA conjugation to the C-terminus of GLP-1 via Michael addition could be used to generate GLP-1 variants with enhanced glucose-lowering activity and prolonged plasma half-life in vivo.
- Published
- 2021
30. Recombinant Peptide Production Platform Coupled with Site-Specific Albumin Conjugation Enables a Convenient Production of Long-Acting Therapeutic Peptide
- Author
-
Kiyoon Min, Jinhwan Cho, Junyong Park, Giyoong Tae, Jihyoun Seong, Young S. Hahn, Mijeong Bak, and Inchan Kwon
- Subjects
Pharmaceutical Science ,lcsh:RS1-441 ,Peptide ,010402 general chemistry ,01 natural sciences ,Article ,albumin conjugation ,law.invention ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,serum half-life extension ,In vivo ,law ,Recombinant peptide ,medicine ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,Albumin ,recombinant therapeutic peptide ,Human serum albumin ,In vitro ,0104 chemical sciences ,Long acting ,Biochemistry ,Recombinant DNA ,medicine.drug - Abstract
The number of therapeutic peptides for human treatment is growing rapidly. However, their development faces two major issues: the poor yield of large peptides from conventional solid-phase synthesis, and the intrinsically short serum half-life of peptides. To address these issues, we investigated a platform for the production of a recombinant therapeutic peptide with an extended serum half-life involving the site-specific conjugation of human serum albumin (HSA). HSA has an exceptionally long serum half-life and can be used to extend the serum half-lives of therapeutic proteins and peptides. We used glucagon-like-peptide 1 (GLP-1) as a model peptide in the present study. A &ldquo, clickable&rdquo, non-natural amino acid&mdash, p-azido-l-phenylalanine (AzF)&mdash, was incorporated into three specific sites (V16, Y19, and F28) of a GLP-1 variant, followed by conjugation with HSA through strain-promoted azide&ndash, alkyne cycloaddition. All three HSA-conjugated GLP-1 variants (GLP1_16HSA, GLP1_19HSA, and GLP1_28HSA) exhibited comparable serum half-lives in vivo. However, the three GLP1_HSA variants had different in vitro biological activities and in vivo glucose-lowering effects, demonstrating the importance of site-specific HSA conjugation. The platform described herein could be used to develop other therapeutic peptides with extended serum half-lives.
- Published
- 2020
31. Repeated Recovery of Rare Earth Elements Using a Highly Selective and Thermo‐Responsive Genetically Encoded Polypeptide (Adv. Funct. Mater. 13/2022)
- Author
-
Zohaib Hussain, Seoungkyun Kim, Jinhwan Cho, Gyudae Sim, Youngjune Park, and Inchan Kwon
- Subjects
Biomaterials ,Electrochemistry ,Condensed Matter Physics ,Electronic, Optical and Magnetic Materials - Published
- 2022
- Full Text
- View/download PDF
32. Repeated Recovery of Rare Earth Elements Using a Highly Selective and Thermo‐Responsive Genetically Encoded Polypeptide
- Author
-
Zohaib Hussain, Seoungkyun Kim, Jinhwan Cho, Gyudae Sim, Youngjune Park, and Inchan Kwon
- Subjects
Biomaterials ,Electrochemistry ,Condensed Matter Physics ,Electronic, Optical and Magnetic Materials - Published
- 2021
- Full Text
- View/download PDF
33. Efficient loading of ophthalmic drugs with poor loadability into contact lenses using functional comonomers
- Author
-
Nohwook Lee, Inchan Kwon, and Dasom Lee
- Subjects
Drug ,Ofloxacin ,genetic structures ,Polymers ,media_common.quotation_subject ,Biomedical Engineering ,Administration, Ophthalmic ,02 engineering and technology ,Methacrylate ,Ophthalmic drugs ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Delivery Systems ,0302 clinical medicine ,law ,medicine ,General Materials Science ,media_common ,Acrylic acid ,Hydrogels ,Neomycin ,Contact Lenses, Hydrophilic ,021001 nanoscience & nanotechnology ,eye diseases ,Anti-Bacterial Agents ,Lens (optics) ,Drug Liberation ,chemistry ,Methacrylic acid ,Delivery efficiency ,030221 ophthalmology & optometry ,sense organs ,0210 nano-technology ,medicine.drug ,Biomedical engineering - Abstract
Ophthalmic formulations have classically been administered to eyes through eye drops, which have poor delivery efficiency and require frequent instillation. As a means of overcoming these drawbacks, ocular-drug delivery via therapeutic contact lenses has caused great interest. A simple way to prepare therapeutic contact lenses is to immerse lenses in concentrated drug solution. However, not all ocular drugs can be efficiently loaded into contact lenses by a simple soaking. In particular, our previous study showed that two antibacterial ocular drugs, ofloxacin (OFX) and neomycin (NEO), were poorly loaded to poly(2-hydroxyethyl methacrylate) (pHEMA)-based contact lenses. Herein, we investigated whether alteration of lens composition using several negatively charged comonomers can enhance loading of ocular drugs with poor loadability (OFX and NEO). As comonomers, methacrylic acid (MAA), acrylic acid (AA), and 4-methyl-4-pentenoic acid (MPA) were used, generating p(HEMA-co-MAA), p(HEMA-co-AA), and p(HEMA-co-MPA) hydrogel-based contact lenses, respectively. Contact lenses containing comonomers exhibited an increase in loading of OFX and NEO. In particular, compared with pHEMA contact lenses, contact lenses containing 2.5 mol% AA exhibited enhanced loading of OFX and NEO, 18 and 53 times, respectively. Charge interactions between comonomers and the drug were considered primary factors in the substantial increase in drug loading.
- Published
- 2018
- Full Text
- View/download PDF
34. Modified hydrogels based on poly(2-hydroxyethyl methacrylate) (pHEMA) with higher surface wettability and mechanical properties
- Author
-
Mincheol Chang, Jihye Jang, Eunsun Seo, Yang-Il Huh, Inchan Kwon, Jung-Hyun Lee, Ji Hoon Park, Santosh Kumar, and Jae-Suk Lee
- Subjects
chemistry.chemical_classification ,Glycidyl methacrylate ,Materials science ,Polymers and Plastics ,General Chemical Engineering ,Comonomer ,Organic Chemistry ,Radical polymerization ,02 engineering and technology ,Polymer ,010402 general chemistry ,021001 nanoscience & nanotechnology ,Methacrylate ,01 natural sciences ,0104 chemical sciences ,Contact angle ,chemistry.chemical_compound ,chemistry ,Chemical engineering ,Methacrylic acid ,Polymer chemistry ,Self-healing hydrogels ,Materials Chemistry ,0210 nano-technology - Abstract
Soft contact lenses made with poly(2-hydroxyethyl methacrylate) (pHEMA) hydrogels modified with various comonomers such as N-vinyl pyrrolidone, methacrylic acid, glycidyl methacrylate, and glycerol monomethacrylate were prepared to investigate the effect of adding the comonomer on the water content, surface wettability, and tensile modulus. These polymers were synthesized by the free radical polymerization of 2-hydroxyethyl methacrylate (HEMA) with the comonomers in the presence of divinyl benzene used as the crosslinker and azobisisobutyronitrile as the initiator. The chemical structure and transmittance of the hydrogels were analyzed by FTIR and UV/Vis spectrophotometers. The surface wettability and tensile modulus were also studied by measuring the contact angle and tensile modulus with a universal testing machine (UTM). Regarding the properties of water in the hydrogels, the ratio between free to bound water was investigated using differential scanning calorimetry (DSC). As the concentration of crosslinker in the hydrogels increases, the tensile strength also increases, whereas the internal water content and contact angle decrease. The effect of the comonomer composition of the hydrogels was also investigated to optimize the various properties of these comonomers for soft contact lenses.
- Published
- 2017
- Full Text
- View/download PDF
35. Bioconjugation and Active Site Design of Enzymes Using Non-natural Amino Acids
- Author
-
Byungseop Yang and Inchan Kwon
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Enzyme complex ,Bioconjugation ,biology ,General Chemical Engineering ,Active site ,General Chemistry ,010402 general chemistry ,Formate dehydrogenase ,01 natural sciences ,Industrial and Manufacturing Engineering ,0104 chemical sciences ,Amino acid ,03 medical and health sciences ,030104 developmental biology ,Enzyme ,chemistry ,Biochemistry ,Dihydrofolate reductase ,biology.protein ,Click chemistry - Abstract
Enzymes are biocatalysts that play key roles in diverse chemical reactions in living organisms and industrial conversion processes generating value-added products. Since wild-type enzymes obtained from nature are normally not optimal for various applications, enzyme engineering is usually required for enhanced or new properties. Site-specific incorporation of a non-natural amino acid became a powerful protein-engineering tool. In this short review, we briefly summarize our contribution to enzyme complex formation and active site design of enzymes using the technique of site-specific incorporation of a non-natural amino acid. First, site-specific incorporation of a non-natural amino acid at a permissive site of an enzyme led to bioconjugation to other molecules without compromising critical properties. Murine dihydrofolate reductase (mDHFR) was site-specifically conjugated to a biotin via click chemistry to achieve site-specific immobilization. Similarly, formate dehydrogenase (FDH) was site-specifically c...
- Published
- 2017
- Full Text
- View/download PDF
36. Enhanced production of Dopa-incorporated mussel adhesive protein using engineered translational machineries
- Author
-
Hyung Joon Cha, Byeongseon Yang, Ye Seul Jeong, Jihyoun Seong, Mincheol Shin, Inchan Kwon, and Byungseop Yang
- Subjects
Methanococcus ,Bioengineering ,medicine.disease_cause ,Protein Engineering ,Applied Microbiology and Biotechnology ,In vivo ,medicine ,Escherichia coli ,Animals ,Tyrosine ,Codon ,chemistry.chemical_classification ,biology ,Proteins ,Mussel ,biology.organism_classification ,Amino acid ,Dihydroxyphenylalanine ,chemistry ,Biochemistry ,Mollusca ,Yield (chemistry) ,Protein Biosynthesis ,Transfer RNA ,Methanocaldococcus ,Biotechnology - Abstract
Mussel adhesive proteins (MAPs) have great potential as bioglues, particularly in wet conditions. Although in vivo residue-specific incorporation of 3,4-dihydroxyphenylalanine (Dopa) in tyrosine-auxotrophic Escherichia coli cells allows for production of Dopa-incorporated bioengineered MAPs (dMAPs), the low production yield hinders the practical application of dMAPs. This low production yield of dMAPs is due to low translational activity of a noncanonical amino acid, Dopa, in E. coli cells. Herein, to enhance the production yield of dMAPs, we investigated the coexpression of Dopa-recognizing tyrosyl-tRNA synthetases (TyrRSs). To use the Dopa-specific Methanococcus jannaschii TyrRS (MjTyrRS-Dopa), we altered the anticodon of tyrosyl-tRNA amber suppressor into AUA (MjtRNATyr AUA ) to recognize a tyrosine codon (AUA). Co-overexpression of MjTyrRS-Dopa and MjtRNATyr AUA increased the production yield of Dopa-incorporated MAP foot protein type 3 (dfp-3) by 57%. Similarly, overexpression of E. coli TyrRS (EcTyrRS) led to a 72% higher production yield of dfp-3. Even with coexpression of Dopa-recognizing TyrRSs, dfp-3 has a high Dopa incorporation yield (over 90%) compared to ones prepared without TyrRS coexpression.
- Published
- 2019
37. Improving cancer therapy through the nanomaterials-assisted alleviation of hypoxia
- Author
-
Inchan Kwon, Giyoong Tae, and Abhishek Sahu
- Subjects
Biophysics ,Cancer therapy ,Bioengineering ,02 engineering and technology ,Partial oxygen ,Metastasis ,Biomaterials ,03 medical and health sciences ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,Hypoxia ,030304 developmental biology ,0303 health sciences ,Oxygen supply ,Tumor microenvironment ,Tumor hypoxia ,business.industry ,Hydrogen Peroxide ,Hypoxia (medical) ,021001 nanoscience & nanotechnology ,medicine.disease ,Nanostructures ,Mechanics of Materials ,Ceramics and Composites ,Cancer research ,Tumor Hypoxia ,medicine.symptom ,0210 nano-technology ,Early phase ,business - Abstract
Hypoxia, resulting from the imbalance between oxygen supply and consumption is a critical component of the tumor microenvironment. It has a paramount impact on cancer growth, metastasis and has long been known as a major obstacle for cancer therapy. However, none of the clinically approved anticancer therapeutics currently available for human use directly tackles this problem. Previous clinical trials of targeting tumor hypoxia with bioreductive prodrugs have failed to demonstrate satisfactory results. Therefore, new ideas are needed to overcome the hypoxia barrier. The method of modulating hypoxia to improve the therapeutic activity is of great interest but remains a considerable challenge. One of the emerging concepts is to supply or generate oxygen at the tumor site to increase the partial oxygen pressure and thereby reverse the hypoxia and its effects. In this review, we present an overview of the recent progress in the development of novel nanomaterials for the alleviation of hypoxic microenvironment. Two main strategies for hypoxia augmentation, i) direct delivery of O2 into the tumor, and ii) in situ O2 generations in the tumor microenvironment through different methods such as catalytic decomposition of endogenous hydrogen peroxide (H2O2) and light-triggered water splitting are discussed in detail. At present, these emerging nanomaterials are in their early phase and expected to grow rapidly in the coming years. Despite the promising start, there are several challenges needed to overcome for successful clinical translation.
- Published
- 2019
38. Co-delivery of therapeutic protein and catalase-mimic nanoparticle using a biocompatible nanocarrier for enhanced therapeutic effect
- Author
-
Manse Kim, Sukhwan Kim, Seoungkyun Kim, Giyoong Tae, Junyong Park, Secheon Jung, Kiyoon Kwon, and Inchan Kwon
- Subjects
Urate Oxidase ,Metabolite ,Pharmaceutical Science ,Nanoparticle ,Metal Nanoparticles ,02 engineering and technology ,Hyperuricemia ,Poloxamer ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Biomimetic Materials ,Cell Line, Tumor ,Animals ,Humans ,Cytotoxicity ,030304 developmental biology ,0303 health sciences ,Drug Carriers ,Chemistry ,Urate oxidase ,Hydrogen Peroxide ,021001 nanoscience & nanotechnology ,Catalase ,In vitro ,Uric Acid ,Mice, Inbred C57BL ,Biophysics ,Female ,Gold ,Nanocarriers ,0210 nano-technology ,Aspergillus flavus - Abstract
Therapeutic proteins are indispensable in the treatment of various human diseases. Despite the many benefits of therapeutic proteins, they also exhibit diverse side effects. Therefore, reducing unwanted side effects of therapeutic proteins as well as enhancing their therapeutic efficacy are very important in developing therapeutic proteins. Urate oxidase (UOX) is a therapeutic enzyme that catalyzes the conversion of uric acid (UA) into a soluble metabolite, and it is used clinically for the treatment of hyperuricemia. Since UA degradation by UOX generates H2O2 (a cytotoxic side product), UOX was co-delivered with catalase-mimic nanoparticles (AuNPs) using biocompatible pluronic-based nanocarriers (NCs) to effectively reduce H2O2-associated toxicity in cultured cells and to enhance UA degradation efficiency in vivo. Simple temperature-dependent size changes of NCs allowed co-encapsulation of both UOX and AuNPs at a high loading efficiency without compromising critical properties, resulting in efficient modulation of a mixing ratio of UOX and AuNPs encapsulated in NCs. Co-localizing UOX and AuNPs in the NCs led to enhanced UA degradation and H2O2 removal in vitro, leading to a great reduction in H2O2-associated cytotoxicity compared with UOX alone or a free mixture of UOX and AuNPs. Furthermore, we demonstrated that co-delivery of UOX and AuNPs using NCs significantly improves in vivo UA degradation compared to simple co-injection of free UOX and AuNPs. More broadly, we showed that biocompatible pluronic-based nanocarriers can be used to deliver a target therapeutic protein along with its toxicity-eliminating agent in order to reduce side effects and enhance efficacy.
- Published
- 2019
39. Chimeric protein of internally duplicated α-type carbonic anhydrase from Dunaliella species for improved expression and CO2 sequestration
- Author
-
Thi Khoa My Nguyen, Mi-Ran Ki, Seung Pil Pack, Inchan Kwon, and Sung Ho Kim
- Subjects
0106 biological sciences ,0301 basic medicine ,chemistry.chemical_classification ,biology ,Protein Data Bank (RCSB PDB) ,Bioengineering ,Chimeric gene ,Dunaliella ,biology.organism_classification ,01 natural sciences ,Applied Microbiology and Biotechnology ,Biochemistry ,Fusion protein ,Amino acid ,03 medical and health sciences ,030104 developmental biology ,Enzyme ,stomatognathic system ,chemistry ,010608 biotechnology ,Carbonic anhydrase ,biology.protein ,Thermostability - Abstract
An internally duplicated α-type carbonic anhydrase (CA) from Dunaliella species (Dsp-CA) has been expressed as single-domain derivatives, N- and C- half domains. Although both derivatives have structures similar to that of a known CA (PDB ID: 1y7w ), only the C-half domain (Dsp-CA-c) exhibited enzymatic activity, albeit with low solubility. In order to increase the solubility of Dsp-CA-c, its proximal N-terminal amino acids 1–11 were substituted with VSEPHDYNYEK (NT11) of the N-half domain (Dsp-CA-n) which exhibits a high soluble expression yield. The new chimeric gene products, Dsp-nCA-c displayed approximately 2-fold the solubility and activity shown by Dsp-CA-c. Dsp-nCA-c showed increased thermal stability (ΔTm > 10 °C) by multimerization compared to Dsp-CA-c. Finally, the chimeric protein effectively catalyzed the conversion of CO2 into its calcite form in the presence of CaCl2. These findings indicate that the substituted NT11 may contribute to soluble expression and enhanced activity of Dsp-nCA-c and cause multimerization which can confer increased thermostability to Dsp-nCA-c. Therefore, N-terminal engineering can be an effective strategy for improving soluble production yield and thermostabillity of CA without disrupting catalytic activity, and the engineered CA could be usefully employed for the development of an efficient enzymatic CO2 sequestration system.
- Published
- 2016
- Full Text
- View/download PDF
40. Positional effects of hydrophobic non-natural amino acid mutagenesis into the surface region of murine dihydrofolate reductase on enzyme properties
- Author
-
Seung Pil Pack, H. Edward Wong, and Inchan Kwon
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Environmental Engineering ,biology ,Artificial enzyme ,Stereochemistry ,Biomedical Engineering ,Bioengineering ,Protein engineering ,010402 general chemistry ,01 natural sciences ,Enzyme structure ,0104 chemical sciences ,Amino acid ,03 medical and health sciences ,030104 developmental biology ,Enzyme ,chemistry ,Biochemistry ,Dihydrofolate reductase ,biology.protein ,Peptide sequence ,Protein secondary structure ,Biotechnology - Abstract
The ability to incorporate non-natural amino acids during protein biosynthesis has greatly broadened the amino acid sequence space available for protein engineering. The increasing number of non-natural amino acids provides novel sidechain chemistries, structures, and functionalities for engineered proteins including enzymes. Therefore, understanding how non-natural amino acid incorporation (non-natural amino acid mutagenesis) affects protein function is crucial. This study investigates the positional effects of hydrophobic non-natural amino acid mutagenesis of the enzyme surface on the enzyme structure and function with the ultimate goal of identification of permissive sites for non-natural amino acid mutagenesis. A bulky, hydrophobic non-natural amino acid, 3-(2-naphthyl)-alanine (2Nal), was site-specifically incorporated at the solvent-exposed surface of a murine dihydrofolate reductase (mDHFR) enzyme. Incorporation of 2Nal was performed at six solvent-exposed sites (V43, E44, F142, E143, F179, and E180), generating six mDHFR variants. Incorporation of 2Nal into F142 or F179 did not exhibit any substantial change in the secondary structure and catalytic activities, whereas 2Nal incorporation at hydrophilic Glu sites significantly changed the secondary structure and catalytic activities. Such different responses upon 2Nal incorporation at hydrophobic and hydrophilic residues were likely due to the structural changes caused by the hydrophobicity change upon mutation. A non-conservative mutation of V43 with 2Nal significantly reduced the catalytic activities suggesting that a substantial size change of sidechain also causes the structural changes. These results suggest that even hydrophobic, bulky non-natural amino acids can be incorporated at the enzyme surface without compromising the enzymatic activities.
- Published
- 2016
- Full Text
- View/download PDF
41. Site-Specific Albumination as an Alternative to PEGylation for the Enhanced Serum Half-Life in Vivo
- Author
-
Giyoong Tae, Jong Chul Kim, Inchan Kwon, Sung In Lim, and Byungseop Yang
- Subjects
Male ,0301 basic medicine ,Polymers and Plastics ,Phenylalanine ,Green Fluorescent Proteins ,Serum albumin ,Bioengineering ,macromolecular substances ,Polyethylene glycol ,Polyethylene Glycols ,law.invention ,Biomaterials ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,law ,PEG ratio ,Materials Chemistry ,medicine ,Animals ,Humans ,Serum Albumin ,Mice, Inbred BALB C ,biology ,Extender ,technology, industry, and agriculture ,Human serum albumin ,030104 developmental biology ,chemistry ,Biochemistry ,PEGylation ,biology.protein ,Target protein ,Half-Life ,medicine.drug ,Conjugate - Abstract
Polyethylene glycol (PEG) has been widely used as a serum half-life extender of therapeutic proteins. However, due to immune responses and low degradability of PEG, developing serum half-life extender alternatives to PEG is required. Human serum albumin (HSA) has several beneficial features as a serum half-life extender, including a very long serum half-life, good degradability, and low immune responses. In order to further evaluate the efficacy of HSA, we compared the extent of serum half-life extension of a target protein, superfolder green fluorescent protein (sfGFP), upon HSA conjugation with PEG conjugation side-by-side. Combination of site-specific incorporation of p-azido-l-phenylalanine into sfGFP and copper-free click chemistry achieved the site-specific conjugation of a single HSA, 20 kDa PEG, or 30 kDa PEG to sfGFP. These sfGFP conjugates exhibited the fluorescence comparable to or even greater than that of wild-type sfGFP (sfGFP-WT). In mice, HSA-conjugation to sfGFP extended the serum half-life 9.0 times compared to that of unmodified sfGFP, which is comparable to those of PEG-conjugated sfGFPs (7.3 times for 20 kDa PEG and 9.5 times for 30 kDa PEG). These results clearly demonstrated that HSA was as effective as PEG in extending the serum half-life of a target protein. Therefore, with the additional favorable features, HSA is a good serum half-life extender of a (therapeutic) protein as an alternative to PEG.
- Published
- 2016
- Full Text
- View/download PDF
42. Oral Triphenylmethane Food Dye Analog, Brilliant Blue G, Prevents Neuronal Loss in APPSwDI/NOS2-/- Mouse Model
- Author
-
Inchan Kwon, Alev Erisir, and Jacob A. Irwin
- Subjects
0301 basic medicine ,Genetically modified mouse ,Pathology ,medicine.medical_specialty ,Transgene ,Drug Evaluation, Preclinical ,Administration, Oral ,Mice, Transgenic ,amyloid-β ,Pharmacology ,Biology ,Hippocampal formation ,010402 general chemistry ,01 natural sciences ,Article ,Random Allocation ,intracellular amyloid-β ,03 medical and health sciences ,Alzheimer Disease ,Oral administration ,Rosaniline Dyes ,medicine ,Animals ,Alzheimer’s Disease ,Cytotoxicity ,Neurons ,Amyloid beta-Peptides ,Cell Death ,Weight change ,Brain ,blood-brain barrier ,In vitro ,0104 chemical sciences ,Mice, Inbred C57BL ,Disease Models, Animal ,Neuroprotective Agents ,030104 developmental biology ,Neurology ,triphenylmethane dye ,Female ,neuronal loss ,Neurology (clinical) ,Intracellular - Abstract
Reducing amyloid-β (Aβ) accumulation is a promising strategy for developing Alzheimer’s Disease (AD) therapeutics. We recently reported that a triphenylmethane food dye analog, Brilliant Blue G (BBG), is a dose-dependent modulator of in vitro amyloid-β aggregation and cytotoxicity in cell-based assays. Following up on this recent work, we sought to further evaluate this novel modulator in a therapeutically-relevant AD transgenic mouse model. BBG was orally administered to APPSwDI/NOS2-/- mice for three months in order to assess its biocompatibility, its permeability across the blood-brain barrier, and its efficacy at rescuing AD pathology. The results showed that BBG was well-tolerated, caused no significant weight change/unusual behavior, and was able to significantly cross the AD blood-brain barrier in APPSwDI/NOS2-/- mice. Immunohistochemical and electron microscopic analysis of the brain sections revealed that BBG was able to significantly prevent neuronal loss and reduce intracellular APP/Aβ in hippocampal neurons. This is the first report of 1) the effect of Brilliant Blue G on neuronal loss in a transgenic animal model of AD, 2) oral administration of BBG to affect a protein conformation/aggregation disease, and 3) electron microscopic ultrastructural analysis of AD pathology in APPSwDI/NOS2-/- mice.
- Published
- 2016
- Full Text
- View/download PDF
43. Bioorthogonal Modification of Proteins Using Genetically Encoded Non-Natural Amino Acids
- Author
-
Sung Lim and Inchan Kwon
- Subjects
Organic Chemistry - Published
- 2016
- Full Text
- View/download PDF
44. The Minimal Effect of Linker Length for Fatty Acid Conjugation to a Small Protein on the Serum Half-Life Extension
- Author
-
Inchan Kwon, Mi Sun Jin, Junyong Park, Jinhwan Cho, and Giyoong Tae
- Subjects
0301 basic medicine ,Serum albumin ,Medicine (miscellaneous) ,02 engineering and technology ,Conjugated system ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Neonatal Fc receptor ,Binding site ,lcsh:QH301-705.5 ,fatty acid conjugation ,chemistry.chemical_classification ,biology ,Albumin ,Serum half-life extension ,Fatty acid ,021001 nanoscience & nanotechnology ,030104 developmental biology ,lcsh:Biology (General) ,Biochemistry ,chemistry ,FcRn-mediated recycling ,biology.protein ,0210 nano-technology ,Linker ,Conjugate - Abstract
Conjugation of serum albumin or one of its ligands (such as fatty acid) has been an effective strategy to prolong the serum half-lives of drugs via neonatal Fc receptor (FcRn)&ndash, mediated recycling of albumin. So far, fatty acid (FA) has been effective in prolonging the serum half-lives for therapeutic peptides and small proteins, but not for large therapeutic proteins. Very recently, it was reported a large protein conjugated to FA competes with the binding of FcRn with serum albumin, leading to limited serum half-life extension, because primary FA binding sites in serum albumin partially overlap with FcRn binding sites. In order to prevent such competition, longer linkers between FA and the large proteins were required. Herein, we hypothesized that small proteins do not cause substantial competition for FcRn binding to albumin, resulting in the extended serum half-life. Using a small protein (28 kDa), we investigated whether the intramolecular distance in FA-protein conjugate affects the FcRn binding with albumin and serum half-life using linkers with varying lengths. Unlike with the FA-conjugated large protein, all FA-conjugated small proteins with different linkers exhibited comparable the FcRn binding to albumin and extended serum half-life.
- Published
- 2020
- Full Text
- View/download PDF
45. Comparative studies of the serum half-life extension of a protein via site-specific conjugation to a species-matched or -mismatched albumin
- Author
-
Giyoong Tae, Jihyoun Seong, Byungseop Yang, Jong Chul Kim, and Inchan Kwon
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Models, Molecular ,Green Fluorescent Proteins ,Biomedical Engineering ,Serum albumin ,Mice, Transgenic ,Plasma protein binding ,Receptors, Fc ,03 medical and health sciences ,Mice ,In vivo ,medicine ,Animals ,Humans ,General Materials Science ,Serum Albumin ,chemistry.chemical_classification ,Mice, Knockout ,Mice, Inbred BALB C ,biology ,Molecular Structure ,Histocompatibility Antigens Class I ,Albumin ,Human serum albumin ,Molecular biology ,Amino acid ,body regions ,030104 developmental biology ,chemistry ,embryonic structures ,biology.protein ,Target protein ,medicine.drug ,Protein Binding - Abstract
Human serum albumin (HSA) has been investigated as a serum half-life extender of therapeutic proteins thanks to its unusually long serum half-life. However, in mice, the serum half-life of a HSA-conjugated protein was much shorter than that of HSA in humans, likely due to the species-dependent nature of albumin–FcRn interactions. Herein, we investigated species-dependent albumin–FcRn interactions using species-matched albumin (mouse serum albumin) and species-mismatched albumin (HSA) in non-transgenic mice. We site-specifically introduced a clickable non-natural amino acid to a target protein followed by conjugation to an albumin species via a hetero-bifunctional linker. Using in vitro binding assays, we showed that both HSA- and MSA-conjugated proteins bound mouse FcRns. Conjugation of HSA led to very limited extension of the serum half-life of sfGFP in mice (16.3 h), compared to that of HSA in transgenic mice harboring an allele of mouse FcRn knock-out and expressing humn FcRn (67 h) reported previously. These results suggest that the FcRn-mediated recycling of HSA is not effective in mice. However, conjugation of mouse serum albumin (MSA) resulted in a serum half-life of sfGFP (27.7 h) comparable to that of MSA in mice (28.8 h). Altogether, our study supported that albumin–FcRn interactions are species dependent in vivo.
- Published
- 2018
46. Expansion of bioorthogonal chemistries towards site-specific polymer–protein conjugation
- Author
-
Secheon Jung and Inchan Kwon
- Subjects
chemistry.chemical_classification ,Polymers and Plastics ,Chemistry ,Organic Chemistry ,A protein ,Bioengineering ,Nanotechnology ,02 engineering and technology ,Polymer ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,Biochemistry ,Combinatorial chemistry ,0104 chemical sciences ,Target protein ,Bioorthogonal chemistry ,0210 nano-technology ,Conjugate - Abstract
Polymer conjugation to proteins has been widely used to improve or expand protein properties. However, polymer conjugation to random sites of a target protein often led to a significant loss of critical protein properties. In order to overcome this, polymer conjugation to specific sites of a protein was developed using the site-specific introduction of non-natural amino acids and bioorthogonal chemistries. This review summarizes the recent advances in bioorthogonal chemistries. As the repertoire of bioorthogonal chemistries available for polymer conjugation is expanding, the site-specific polymer conjugation technique would be a valuable platform technique to design novel protein–polymer conjugates.
- Published
- 2016
- Full Text
- View/download PDF
47. Manipulating the substrate specificity of murine dihydrofolate reductase enzyme using an expanded set of amino acids
- Author
-
Inchan Kwon, Sung In Lim, and Shun Zheng
- Subjects
chemistry.chemical_classification ,Environmental Engineering ,biology ,Artificial enzyme ,Stereochemistry ,Protein design ,Biomedical Engineering ,Active site ,Bioengineering ,Ligand (biochemistry) ,Amino acid ,Enzyme ,chemistry ,Biochemistry ,Docking (molecular) ,Dihydrofolate reductase ,biology.protein ,Biotechnology - Abstract
As the number of reactions requiring biotransformation continues to grow, manipulating the enzyme substrate specificity becomes very important. Complementary to conventional enzyme engineering techniques based on only natural amino acids, we hypothesized that the site-specific incorporation of a non-natural amino acid in vivo into an enzyme can be used to re-design the active site for the altered substrate specificity. To test our hypothesis, we introduced the non-natural amino acid L -2-naphthylalanine (2Nal) into the active site of the model enzyme murine dihydrofolate reductase (mDHFR). We explored whether the substrate specificity of the enzyme could be switched from the good substrate dihydrofolate (DHF) to the poor substrate folate (FOL). We used two protein design programs (RosettaLigand and RosettaDesign) to calculate ligand docking and conformational stability, respectively, for the evaluation of multiples sites in the mDHFR. From the calculations, position 31 was predicted as an optimal 2Nal incorporation site. One mDHFR variant containing 2Nal at position 31 (mDHFR 2Nal31 ) was expressed in the Escherichia coli expression host cells equipped with the engineered yeast phenylalanyl-tRNA and phenylalanyl-tRNA synthetase pair. As expected, the kinetic assays of purified mDHFR variant revealed that mDHFR 2Nal31 has the enhanced binding affinity toward FOL and also exhibits 7.6-fold enhanced catalytic efficiency of FOL over DHF compared to mDHFR WT .
- Published
- 2015
- Full Text
- View/download PDF
48. Bioconjugation of therapeutic proteins and enzymes using the expanded set of genetically encoded amino acids
- Author
-
Sung In Lim and Inchan Kwon
- Subjects
chemistry.chemical_classification ,Bioconjugation ,010405 organic chemistry ,Biomolecule ,Proteins ,General Medicine ,Protein engineering ,010402 general chemistry ,Genetic code ,01 natural sciences ,Applied Microbiology and Biotechnology ,0104 chemical sciences ,Amino acid ,Enzyme ,chemistry ,Biochemistry ,Genetic Code ,Animals ,Humans ,Amino Acids ,Bioorthogonal chemistry ,Expanded genetic code ,Biotechnology - Abstract
The last decade has witnessed striking progress in the development of bioorthogonal reactions that are strictly directed towards intended sites in biomolecules while avoiding interference by a number of physical and chemical factors in biological environment. Efforts to exploit bioorthogonal reactions in protein conjugation have led to the evolution of protein translational machineries and the expansion of genetic codes that systematically incorporate a range of non-natural amino acids containing bioorthogonal groups into recombinant proteins in a site-specific manner. Chemoselective conjugation of proteins has begun to find valuable applications to previously inaccessible problems. In this review, we describe bioorthogonal reactions useful for protein conjugation, and biosynthetic methods that produce proteins amenable to those reactions through an expanded genetic code. We then provide key examples in which novel protein conjugates, generated by the genetic incorporation of a non-natural amino acid and the chemoselective reactions, address unmet needs in protein therapeutics and enzyme engineering.
- Published
- 2015
- Full Text
- View/download PDF
49. Site-specific albumination of a therapeutic protein with multi-subunit to prolong activity in vivo
- Author
-
Young S. Hahn, Sung In Lim, and Inchan Kwon
- Subjects
Urate Oxidase ,Recombinant Fusion Proteins ,Pharmaceutical Science ,Serum Albumin, Human ,Protein Engineering ,Article ,Fungal Proteins ,Drug Stability ,In vivo ,Enzyme Stability ,medicine ,Animals ,Serum Albumin ,chemistry.chemical_classification ,Albumin ,Urate oxidase ,Human serum albumin ,Amino acid ,Mice, Inbred C57BL ,chemistry ,Biochemistry ,Area Under Curve ,Injections, Intravenous ,Female ,Target protein ,Aspergillus flavus ,Half-Life ,Homotetramer ,Conjugate ,medicine.drug - Abstract
Albumin fusion/conjugation (albumination) has been an effective method to prolong in vivo half-life of therapeutic proteins. However, its broader application to proteins with complex folding pathway or multi-subunit is restricted by incorrect folding, poor expression, heterogeneity, and loss of native activity of the proteins linked to albumin. We hypothesized that the site-specific conjugation of albumin to a permissive site of a target protein will expand the utilities of albumin as a therapeutic activity extender to proteins with a complex structure. We show here the genetic incorporation of a non-natural amino acid (NNAA) followed by chemoselective albumin conjugation to prolong therapeutic activity in vivo. Urate oxidase (Uox), a therapeutic enzyme for treatment of hyperuricemia, is a homotetramer with multiple surface lysines, limiting conventional approaches for albumination. Incorporation of p-azido-l-phenylalanine into two predetermined positions of Uox allowed site-specific linkage of dibenzocyclooctyne-derivatized human serum albumin (HSA) through strain-promoted azide-alkyne cycloaddition (SPAAC). The bio-orthogonality of SPAAC resulted in the production of a chemically well-defined conjugate, Uox-HSA, with a retained enzymatic activity. Uox-HSA had a half-life of 8.8 h in mice, while wild-type Uox had a half-life of 1.3 h. The AUC increased 5.5-fold (1657 vs. 303 mU/mL × h). These results clearly demonstrated that site-specific albumination led to the prolonged enzymatic activity of Uox in vivo. Site-specific albumination enabled by NNAA incorporation and orthogonal chemistry demonstrates its promise for the development of long-acting protein therapeutics with high potency and safety.
- Published
- 2015
- Full Text
- View/download PDF
50. A Chemical Chaperone-Based Drug Candidate is Effective in a Mouse Model of Amyotrophic Lateral Sclerosis (ALS)
- Author
-
Olga Viskind, Arie Gruzman, Hanoch Senderowitz, Susanne Petri, Daniel Offen, Omer Green, Inchan Kwon, Yael Barhum, Simpson Gregoire, Ilana Zaks, Tom Shani, Hugo E. Gottlieb, Tali Ben-Zur, Adrian Israelson, Sebastian Böselt, Tamar Getter, and Moran Shubely
- Subjects
Mice, Transgenic ,Pharmacology ,Biochemistry ,Mice ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Animals ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Amyotrophic lateral sclerosis ,Cells, Cultured ,Dose-Response Relationship, Drug ,Molecular Structure ,Amyotrophic Lateral Sclerosis ,Organic Chemistry ,medicine.disease ,Amides ,Small molecule ,In vitro ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Drug development ,Molecular Medicine ,Protein folding ,Chemical chaperone ,Lead compound ,Astrocyte - Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the selective death of motor neurons and skeletal muscle atrophy. The majority of ALS cases are acquired spontaneously, with inherited disease accounting for only 10 % of all cases. Recent studies provide compelling evidence that aggregates of misfolded proteins underlie both types of ALS. Small molecules such as artificial chaperones can prevent or even reverse the aggregation of proteins associated with various human diseases. However, their very high active concentration (micromolar range) severely limits their utility as drugs. We synthesized several ester and amide derivatives of chemical chaperones. The lead compound 14, 3-((5-((4,6-dimethylpyridin-2-yl)methoxy)-5-oxopentanoyl)oxy)-N,N-dimethylpropan-1-amine oxide shows, in the micromolar concentration range, both neuronal and astrocyte protective effects in vitro; at daily doses of 10 mg kg(-1) 14 improved the neurological functions and delayed body weight loss in ALS mice. Members of this new chemical chaperone derivative class are strong candidates for the development of new drugs for ALS patients.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.