“Tag and Modify” Protein Conjugation with Dynamic Covalent ChemistryClick to copy article linkArticle link copied!
- Maksymilian Marek ZegotaMaksymilian Marek ZegotaMax-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, GermanyInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanyMore by Maksymilian Marek Zegota
- Tao WangTao WangInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanySchool of Materials Science and Engineering, Southwest Jiaotong University, 610031 Chengdu, P.R. ChinaMore by Tao Wang
- Christiane SeidlerChristiane SeidlerMax-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, GermanyInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanyMore by Christiane Seidler
- David Yuen Wah NgDavid Yuen Wah NgMax-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, GermanyInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanyMore by David Yuen Wah Ng
- Seah Ling Kuan*Seah Ling Kuan*E-mail: [email protected]Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, GermanyInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanyMore by Seah Ling Kuan
- Tanja Weil*Tanja Weil*E-mail: [email protected]Max-Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, GermanyInstitute of Inorganic Chemistry I, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, GermanyMore by Tanja Weil
Abstract
The development of small protein tags that exhibit bioorthogonality, bond stability, and reversibility, as well as biocompatibility, holds great promise for applications in cellular environments enabling controlled drug delivery or for the construction of dynamic protein complexes in biological environments. Herein, we report the first application of dynamic covalent chemistry both for purification and for reversible assembly of protein conjugates using interactions of boronic acid with diols and salicylhydroxamates. Incorporation of the boronic acid (BA) tag was performed in a site-selective fashion by applying disulfide rebridging strategy. As an example, a model protein enzyme (lysozyme) was modified with the BA tag and purified using carbohydrate-based column chromatography. Subsequent dynamic covalent “click-like” bioconjugation with a salicylhydroxamate modified fluorescent dye (BODIPY FL) was accomplished while retaining its original enzymatic activity.
Introduction
Results and Discussion
Synthesis and Purification of BA-Tagged Lysozyme
Scheme 1
aa. methacryloyl chloride, Et3N, CH2Cl2, 99%; b. 1. I2, sodium tosylate, CH2Cl2; 2. Et3N, EA, 52%; c. TFA, CH2Cl2, 99%; d. HBTU, DIEA, 3-aminophenylboronic acid hydrochloride, DMF, 65%.
Scheme 2
Figure 1
Figure 1. FPLC chromatogram of the reaction mixture with visible separation of lysozyme, modified lysozyme, and a higher molecular weight side product (retention volume = 19.82 mL).
Figure 2
Figure 2. (a) MALDI-TOF-MS interpretation of BA-lysozyme digest. Peak corresponding to modified fragment was selected for MSMS. 2,5-Dihydrobenzoic acid (DHB) was used as a matrix. (b) Interpretation of BA modified fragment’s MSMS spectrum. To simplify, only a, b, and y ion series for 6–13 fragment are annotated. Annotations for a-17, b-17 ion series and 126–128 fragment are included in the Supporting Information.
Figure 3
Figure 3. Rate of decrease in absorbance (slope) is proportional to lysozyme activity. Clearance of cell suspension turbidity proceeds with similar pace in case of both BA-modified (orange) and native lysozyme (red). Data are plotted as n = 4 ± standard errors of the mean (SEM).
Reversible Functionalization of BA-Lysozyme
Scheme 3
aa. CuSO4, sodium ascorbate, DMSO, 93%; b. BF3OEt2, MeOH/CH2Cl2, 76%.
Scheme 4
Figure 4
Figure 4. Microscale thermophoresis confirms assembly of BA-lysozyme with SHA-BDP (blue line) and release of SHA-BDP upon acidification (gray dots). No binding was detected for the control (red and green dots). Data are plotted as n = 3 ± SEM.
Conclusion
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.8b00358.
Full experimental procedures and characterization data for new compounds (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This project has received funding from the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement No 675007. M. M. Zegota thanks the Marie Curie International Training Network Protein Conjugates for a research scholarship. The authors are grateful to the Max Planck Society, German National Foundation (Sonderforschungsbereich 1149, Project B11, Sonderforschungsbereich 1297, Project C01) and the Marie Skłodowska-Curie Actions (MSCA) for the financial support.
BA | boronic acid |
BODIPY (BDP) | boron-dipyrromethene |
Kd | dissociation constant |
SHA | salicylhydroxamate. |
References
This article references 53 other publications.
- 1Dozier, J. K. and Distefano, M. D. (2015) Site-Specific PEGylation of Therapeutic Proteins. Int. J. Mol. Sci. 16, 25831– 25864, DOI: 10.3390/ijms161025831Google Scholar1Site-specific pegylation of therapeutic proteinsDozier, Jonathan K.; Distefano, Mark D.International Journal of Molecular Sciences (2015), 16 (10), 25831-25864CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)The use of proteins as therapeutics has a long history and is becoming ever more common in modern medicine. While the no. of protein-based drugs is growing every year, significant problems still remain with their use. Among these problems are rapid degrdn. and excretion from patients, thus requiring frequent dosing, which in turn increases the chances for an immunol. response as well as increasing the cost of therapy. One of the main strategies to alleviate these problems is to link a polyethylene glycol (PEG) group to the protein of interest. This process, called PEGylation, has grown dramatically in recent years resulting in several approved drugs. Installing a single PEG chain at a defined site in a protein is challenging. Recently, there is has been considerable research into various methods for the site-specific PEGylation of proteins. This review seeks to summarize that work and provide background and context for how site-specific PEGylation is performed. After introducing the topic of site-specific PEGylation, recent developments using chem. methods are described. That is followed by a more extensive discussion of bioorthogonal reactions and enzymic labeling.
- 2Krall, N., da Cruz, F. P., Boutureira, O., and Bernardes, G. J. (2016) Site-selective protein-modification chemistry for basic biology and drug development. Nat. Chem. 8, 103– 113, DOI: 10.1038/nchem.2393Google Scholar2Site-selective protein-modification chemistry for basic biology and drug developmentKrall, Nikolaus; da Cruz, Filipa P.; Boutureira, Omar; Bernardes, Goncalo J. L.Nature Chemistry (2016), 8 (2), 103-113CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. Nature has produced intricate machinery to covalently diversify the structure of proteins after their synthesis in the ribosome. In an attempt to mimic nature, chemists have developed a large set of reactions that enable post-expression modification of proteins at pre-detd. sites. These reactions are now used to selectively install particular modifications on proteins for many biol. and therapeutic applications. For example, they provide an opportunity to install post-translational modifications on proteins to det. their exact biol. roles. Labeling of proteins in live cells with fluorescent dyes allows protein uptake and intracellular trafficking to be tracked and also enables physiol. parameters to be measured optically. Through the conjugation of potent cytotoxicants to antibodies, novel anti-cancer drugs with improved efficacy and reduced side effects may be obtained. In this Perspective, we highlight the most exciting current and future applications of chem. site-selective protein modification and consider which hurdles still need to be overcome for more widespread use.
- 3Kuan, S. L., Wang, T., and Weil, T. (2016) Site-Selective Disulfide Modification of Proteins: Expanding Diversity beyond the Proteome. Chem. - Eur. J. 22, 17112– 17129, DOI: 10.1002/chem.201602298Google Scholar3Site-Selective Disulfide Modification of Proteins: Expanding Diversity beyond the ProteomeKuan, Seah Ling; Wang, Tao; Weil, TanjaChemistry - A European Journal (2016), 22 (48), 17112-17129CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The synthetic transformation of polypeptides with mol. accuracy holds great promise for providing functional and structural diversity beyond the proteome. Consequently, the last decade has seen an exponential growth of site-directed chem. to install addnl. features into peptides and proteins even inside living cells. The disulfide rebridging strategy has emerged as a powerful tool for site-selective modifications since most proteins contain disulfide bonds. In this Review, we present the chem. design, advantages and limitations of the disulfide rebridging reagents, while summarizing their relevance for synthetic customization of functional protein bioconjugates, as well as the resultant impact and advancement for biomedical applications.
- 4Cho, H., Daniel, T., Buechler, Y. J., Litzinger, D. C., Maio, Z., Putnam, A.-M. H., Kraynov, V. S., Sim, B.-C., Bussell, S., Javahishvili, T., Kaphle, S., Viramontes, G., Ong, M., Chu, S., Becky, G. C., Lieu, R., Knudsen, N., Castiglioni, P., Norman, T. C., Axelrod, D. W., Hoffman, A. R., Schultz, P. G., DiMarchi, R. D., and Kimmel, B. E. (2011) Optimized clinical performance of growth hormone with an expanded genetic code. Proc. Natl. Acad. Sci. U. S. A. 108, 9060– 5, DOI: 10.1073/pnas.1100387108Google Scholar4Optimized clinical performance of growth hormone with an expanded genetic codeCho, Ho; Daniel, Tom; Buechler, Ying Ji; Litzinger, David C.; Maio, Zhenwei; Putnam, Anna-Maria Hays; Kraynov, Vadim S.; Sim, Bee-Cheng; Bussell, Stuart; Javahishvili, Tsotne; Kaphle, Sami; Viramontes, Guillermo; Ong, Mike; Chu, Stephanie; Becky, G. C.; Lieu, Ricky; Knudsen, Nick; Castiglioni, Paola; Norman, Thea C.; Axelrod, Douglas W.; Hoffman, Andrew R.; Schultz, Peter G.; DiMarchi, Richard D.; Kimmel, Bruce E.Proceedings of the National Academy of Sciences of the United States of America (2011), 108 (22), 9060-9065, S9060/1-S9060/3CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The ribosomal incorporation of nonnative amino acids into polypeptides in living cells provides the opportunity to endow therapeutic proteins with unique pharmacol. properties. We report here the first clin. study of a biosynthetic protein produced using an expanded genetic code. Incorporation of p-acetylphenylalanine (pAcF) at distinct locations in human growth hormone (hGH) allowed site-specific conjugation with polyethylene glycol (PEG) to produce homogeneous hGH variants. A mono-PEGylated mutant hGH modified at residue 35 demonstrated favorable pharmacodynamic properties in GH-deficient rats. Clin. studies in GH-deficient adults demonstrated efficacy and safety comparable to native human growth hormone therapy but with increased potency and reduced injection frequency. This example illustrates the utility of nonnative amino acids to optimize protein therapeutics in an analogous fashion to the use of medicinal chem. to optimize conventional natural products, low mol. wt. drugs, and peptides.
- 5Elliott, T. S., Townsley, F. M., Bianco, A., Ernst, R. J., Sachdeva, A., Elsässer, S. J., Davis, L., Lang, K., Pisa, R., Greiss, S., Lilley, K. S., and Chin, J. W. (2014) Proteome labeling and protein identification in specific tissues and at specific developmental stages in an animal. Nat. Biotechnol. 32, 465– 472, DOI: 10.1038/nbt.2860Google Scholar5Proteome labeling and protein identification in specific tissues and at specific developmental stages in an animalElliott, Thomas S.; Townsley, Fiona M.; Bianco, Ambra; Ernst, Russell J.; Sachdeva, Amit; Elsaesser, Simon J.; Davis, Lloyd; Lang, Kathrin; Pisa, Rudolf; Greiss, Sebastian; Lilley, Kathryn S.; Chin, Jason W.Nature Biotechnology (2014), 32 (5), 465-472CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Identifying the proteins synthesized at specific times in cells of interest in an animal will facilitate the study of cellular functions and dynamic processes. Here we introduce stochastic orthogonal recoding of translation with chemoselective modification (SORT-M) to address this challenge. SORT-M involves modifying cells to express an orthogonal aminoacyl-tRNA synthetase/tRNA pair to enable the incorporation of chem. modifiable analogs of amino acids at diverse sense codons in cells in rich media. We apply SORT-M to Drosophila melanogaster fed std. food to label and image proteins in specific tissues at precise developmental stages with diverse chemistries, including cyclopropene-tetrazine inverse electron demand Diels-Alder cycloaddn. reactions. We also use SORT-M to identify proteins synthesized in germ cells of the fly ovary without dissection. SORT-M will facilitate the definition of proteins synthesized in specific sets of cells to study development, and learning and memory in flies, and may be extended to other animals.
- 6Boutureira, O. and Bernardes, G. J. L. (2015) Advances in chemical protein modification. Chem. Rev. 115, 2174– 2195, DOI: 10.1021/cr500399pGoogle Scholar6Advances in Chemical Protein ModificationBoutureira, Omar; Bernardes, Goncalo J. L.Chemical Reviews (Washington, DC, United States) (2015), 115 (5), 2174-2195CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Transition metal-free and -mediated approaches are covered.
- 7Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S., Lu, Y., Meng, Y. G., Ng, C., Yang, J., Lee, C. C., Duenas, E., Gorrell, J., Katta, V., Kim, A., McDorman, K., Flagella, K., Venook, R., Ross, S., Spencer, S. D., Lee Wong, W., Lowman, H. B., Vandlen, R., Sliwkowski, M. X., Scheller, R. H., Polakis, P., and Mallet, W. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26, 925– 932, DOI: 10.1038/nbt.1480Google Scholar7Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic indexJunutula, Jagath R.; Raab, Helga; Clark, Suzanna; Bhakta, Sunil; Leipold, Douglas D.; Weir, Sylvia; Chen, Yvonne; Simpson, Michelle; Tsai, Siao Ping; Dennis, Mark S.; Lu, Yanmei; Meng, Y. Gloria; Ng, Carl; Yang, Jihong; Lee, Chien C.; Duenas, Eileen; Gorrell, Jeffrey; Katta, Viswanatham; Kim, Amy; McDorman, Kevin; Flagella, Kelly; Venook, Rayna; Ross, Sarajane; Spencer, Susan D.; Wong, Wai Lee; Lowman, Henry B.; Vandlen, Richard; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Mallet, WilliamNature Biotechnology (2008), 26 (8), 925-932CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogeneous conjugates with relatively narrow therapeutic index (max. tolerated dose/curative dose). Using leads from the authors' previously described phage display-based method to predict suitable conjugation sites, the authors engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb Ig folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepd. by conventional approaches. The favorable in vivo properties of the near-homogeneous compn. of this conjugate suggest that this strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.
- 8Stephan, J.-P., Chan, P., Lee, C., Nelson, C., Elliott, J. M., Bechtel, C., Raab, H., Xie, D., Akutagawa, J., Baudys, J., Saad, O., Prabhu, S., Wong, W. L. T., Vandlen, R., Jacobson, F., and Ebens, A. (2008) Anti-CD22-MCC-DM1 and MC-MMAF Conjugates: Impact of Assay Format on Pharmacokinetic Parameters Determination. Bioconjugate Chem. 19, 1673– 1683, DOI: 10.1021/bc800059tGoogle Scholar8Anti-CD22-MCC-DM1 and MC-MMAF Conjugates: Impact of Assay Format on Pharmacokinetic Parameters DeterminationStephan, Jean-Philippe; Chan, Pamela; Lee, Chien; Nelson, Christopher; Elliott, James Michael; Bechtel, Charity; Raab, Helga; Xie, David; Akutagawa, Jon; Baudys, Jakub; Saad, Ola; Prabhu, Saileta; Wong, Wai Lee T.; Vandlen, Richard; Jacobson, Fred; Ebens, AllenBioconjugate Chemistry (2008), 19 (8), 1673-1683CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)CD22 represents a promising target for antibody-drug conjugate therapy in the context of B cell malignancies since it rapidly internalizes, importing specifically bound antibodies with it. To det. the pharmacokinetic parameters of anti-CD22-MCC-DM1 and MC-MMAF conjugates, various approaches to quantifying total and conjugated antibody were investigated. Although the total antibody assay formats gave similar results for both conjugates, the mouse pharmacokinetic profile for the anti-CD22-MCC-DM1 and MC-MMAF appeared significantly different depending on the conjugated antibody assay format. Since these differences significantly impacted the PK parameters detn., we investigated the effect of the drug/antibody ratio on the total and conjugated antibody quantification using multiple assay formats. Our investigations revealed the limitations of some assay formats to quantify anti-CD22-MCC-DM1 and MC-MMAF with different drug load and in the context of a heterogeneous ADC population highlight the need to carefully plan the assay strategy for the total and conjugated antibody quantification in order to accurately det. the ADC PK parameters.
- 9Chalker, J. M., Bernardes, G. J. L., and Davis, B. G. (2011) A “Tag-and-Modify” Approach to Site-Selective Protein Modification. Acc. Chem. Res. 44, 730– 741, DOI: 10.1021/ar200056qGoogle Scholar9A "Tag-and-Modify" Approach to Site-Selective Protein ModificationChalker, Justin M.; Bernardes, Goncalo J. L.; Davis, Benjamin G.Accounts of Chemical Research (2011), 44 (9), 730-741CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Covalent modification can expand a protein's functional capacity. Fluorescent or radioactive labeling, for instance, allows imaging of a protein in real time. Labeling with an affinity probe enables isolation of target proteins and other interacting mols. At the other end of this functional spectrum, protein structures can be naturally altered by enzymic action. Protein-protein interactions, genetic regulation, and a range of cellular processes are under the purview of these post-translational modifications. The ability of protein chemists to install these covalent addns. selectively has been crit. for elucidating their roles in biol. Frequently the transformations must be applied in a site-specific manner, which demands the most selective chem. In this Account, we discuss the development and application of such chem. in our lab. A centerpiece of our strategy is a "tag-and-modify" approach, which entails sequential installation of a uniquely reactive chem. group into the protein (the "tag") and the selective or specific modification of this group. The chem. tag can be a natural or unnatural amino acid residue. Of the natural residues, cysteine is the most widely used as a tag. Early work in our program focused on selective disulfide formation in the synthesis of glycoproteins. For certain applications, the susceptibility of disulfides to redn. was a limitation and prompted the development of several methods for the synthesis of more stable thioether modifications. The desulfurization of disulfides and conjugate addn. to dehydroalanine are two routes to these modifications. The dehydroalanine tag has since proven useful as a general precursor to many modifications after conjugate addn. of various nucleophiles; phosphorylated, glycosylated, peptidylated, prenylated, and even mimics of methylated and acetylated lysine-contg. proteins are all accessible from dehydroalanine. While cysteine is a useful tag for selective modification, unnatural residues present the opportunity for bio-orthogonal chem. Azide-, arylhalide-, alkyne-, and alkene-contg. amino acids can be incorporated into proteins genetically and can be specifically modified through various transformations. These transformations often rely on metal catalysis. The Cu-catalyzed azide-alkyne addn., Ru-catalyzed olefin metathesis, and Pd-catalyzed cross-coupling are examples of such transformations. In the course of adapting these reactions to protein modification, we learned much about the behavior of these reactions in water, and in some cases entirely new catalysts were developed. Through a combination of these bio-orthogonal transformations from the panel of tag-and-modify reactions, multiple and distinct modifications can be installed on protein surfaces. Multiple modifications are common in natural systems, and synthetic access to these proteins has enabled study of their biol. role. Throughout these investigations, much has been learned in chem. and biol. The demands of selective protein modification have revealed many aspects of reaction mechanisms, which in turn have guided the design of reagents and catalysts that allow their successful deployment in water and in biol. milieu. With this ability to modify proteins, it is now possible to interrogate biol. systems with precision that was not previously possible.
- 10Zhang, X., Lu, W., Kwan, K., Bhattacharyya, D., and Wei, Y. (2017) Dual-Functional-Tag-Facilitated Protein Labeling and Immobilization. ACS Omega 2, 522– 528, DOI: 10.1021/acsomega.6b00512Google ScholarThere is no corresponding record for this reference.
- 11Rybak, J.-N., Scheurer, S. B., Neri, D., and Elia, G. (2004) Purification of biotinylated proteins on streptavidin resin: A protocol for quantitative elution. Proteomics 4, 2296– 2299, DOI: 10.1002/pmic.200300780Google Scholar11Purification of biotinylated proteins on streptavidin resin: A protocol for quantitative elutionRybak, Jascha-N.; Scheurer, Simone B.; Neri, Dario; Elia, GiulianoProteomics (2004), 4 (8), 2296-2299CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)The interaction between streptavidin and biotin is one of the most widely used tools in chem. and biol. However, the release of biotinylated proteins from streptavidin resins remains a major problem, due to the extraordinary stability of this complex. We present a new protocol for the quant. elution of biotinylated proteins from streptavidin Sepharose, featuring harsh elution conditions and competition with free biotin. The usefulness of the method was demonstrated by the quant. recovery of biotinylated proteins from organ homogenates, obtained from mice perfused with a reactive ester deriv. of biotin.
- 12Campos, S. K., Parrott, M. B., and Barry, M. A. (2004) Avidin-based targeting and purification of a protein IX-modified, metabolically biotinylated adenoviral vector. Mol. Ther. 9, 942– 54, DOI: 10.1016/j.ymthe.2004.03.006Google Scholar12Avidin-based targeting and purification of a protein IX-modified, metabolically biotinylated adenoviral vectorCampos, Samuel K.; Parrott, M. Brandon; Barry, Michael A.Molecular Therapy (2004), 9 (6), 942-954CODEN: MTOHCK; ISSN:1525-0016. (Elsevier)While genetic modification of adenoviral vectors can produce vectors with modified tropism, incorporation of targeting peptides/proteins into the structural context of the virion can also result in destruction of ligand targeting or virion integrity. To combat this problem, the authors have developed a versatile targeting system using metabolically biotinylated adenoviral vectors bearing biotinylated fiber proteins. These vectors have been demonstrated to be useful as a platform for avidin-based ligand screening and vector targeting by conjugating biotinylated ligands to the virus using high-affinity tetrameric avidin (Kd = 10-15 M). The biotinylated vector could also be purified by biotin-reversible binding on monomeric avidin (Kd = 10-7 M). In this report, a second metabolically biotinylated adenovirus vector, Ad-IX-BAP, has been engineered by fusing a biotin acceptor peptide (BAP) to the C-terminus of the adenovirus pIX protein. This biotinylated vector displays twice as many biotins and was markedly superior for single-step affinity purifn. on monomeric avidin resin. However, unlike the fiber-biotinylated vector, Ad-IX-BAP failed to retarget to cells with biotinylated antibodies including anti-CD71 against the transferrin receptor. In contrast, Ad-IX-BAP was retargeted if transferrin, the cognate ligand for CD71, was used as a ligand rather than the anti-CD71. This work demonstrates the utility of metabolic biotinylation as a mol. screening tool to assess the utility of different viral capsid proteins for ligand display and the biol. and compatibility of different ligands and receptors for vector targeting applications. These results also demonstrate the utility of the pIX-biotinylated vector as a platform for gentle single-step affinity purifn. of adenoviral vectors.
- 13Bornhorst, J. A. and Falke, J. J. (2000) Purification of proteins using polyhistidine affinity tags. Methods Enzymol. 326, 245– 254, DOI: 10.1016/S0076-6879(00)26058-8Google Scholar13Purification of proteins using polyhistidine affinity tagsBornhorst, Joshua A.; Falke, Joseph J.Methods in Enzymology (2000), 326 (Applications of Chimeric Genes and Hybrid Proteins, Pt. A), 245-254CODEN: MENZAU; ISSN:0076-6879. (Academic Press)A review with 30 refs. The expression and subsequent purifn. of recombinant proteins are widely employed in biochem. studies. A powerful purifn. method involves the use of peptide affinity tags, which are fused to the protein of interest and used to expedite protein purifn. via affinity chromatog. A widely employed method utilizes immobilized metal-affinity chromatog. (IMAC) to purify recombinant proteins contg. a short affinity tag consisting of polyhistidine residues. IMAC is a versatile method that can be utilized to rapidly purify polyhistidine affinity-tagged proteins, resulting in 100-fold enrichments in a single purifn. step. Affinity-tagged protein purities can be achieved at up to 95% purity by IMAC in high yield. Protocols for the method are described in detail. (c) 2000 Academic Press.
- 14Kimple, M. E., Brill, A. L., and Pasker, R. L. (2013) Overview of Affinity Tags for Protein Purification. Curr. Protoc Protein Sci. 73, 608– 616, DOI: 10.1002/0471140864.ps0909s73Google ScholarThere is no corresponding record for this reference.
- 15Fexby, S. and Bü Low, L. (2004) Hydrophobic peptide tags as tools in bioseparation. Trends Biotechnol. 22, 511– 516, DOI: 10.1016/j.tibtech.2004.08.005Google ScholarThere is no corresponding record for this reference.
- 16Jin, Y., Yu, C., Denman, R. J., and Zhang, W. (2013) Recent advances in dynamic covalent chemistry. Chem. Soc. Rev. 42, 6634– 6654, DOI: 10.1039/c3cs60044kGoogle Scholar16Recent advances in dynamic covalent chemistryJin, Yinghua; Yu, Chao; Denman, Ryan J.; Zhang, WeiChemical Society Reviews (2013), 42 (16), 6634-6654CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Dynamic covalent chem. (DCvC) has been strongly integrated into diverse research fields, and has enabled easy access to a variety of combinatorial libraries, 2-D macrocycles, and 3-D mol. cages that target many important applications, such as drug discovery, biotechnol., mol. sepn., light harvesting, etc. DCvC relies on the reversible formation and breaking of rather strong covalent bonding within mols. Therefore it combines the error-correction capability of supramol. chem. and the robustness of covalent bonding. Compared to those supramol. interactions, dynamic covalent reactions usually have slower kinetics and require the assistance of catalysts to achieve rapid equil. Although the scope of dynamic covalent reactions is rapidly expanding, the reversible reactions suitable for DCvC are still very limited. The identification and development of new dynamic reactions and catalysts would be crit. for the further advancement of DCvC. This review covers the recent development of dynamic covalent reactions as well as their applications.
- 17Bartolami, E., Bessin, Y., Gervais, V., Dumy, P., and Ulrich, S. (2015) Dynamic Expression of DNA Complexation with Self-assembled Biomolecular Clusters. Angew. Chem., Int. Ed. 54, 10183– 10187, DOI: 10.1002/anie.201504047Google Scholar17Dynamic Expression of DNA Complexation with Self-assembled Biomolecular ClustersBartolami, Eline; Bessin, Yannick; Gervais, Virginie; Dumy, Pascal; Ulrich, SebastienAngewandte Chemie, International Edition (2015), 54 (35), 10183-10187CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We report herein the implementation of a dynamic covalent chem. approach to the generation of multivalent clusters for DNA recognition. We show that biomol. clusters can be expressed in situ by a programmed self-assembly process using chemoselective ligations. The cationic clusters are shown, by fluorescence displacement assay, gel electrophoresis and isothermal titrn. calorimetry, to effectively complex DNA through multivalent interactions. The reversibility of the ligation was exploited to demonstrate that template effects occur, whereby DNA imposes component selection in order to favor the most active DNA-binding clusters. Furthermore, we show that a chem. effector can be used to trigger DNA release through component exchange reactions.
- 18Reuther, J. F., Dees, J. L., Kolesnichenko, I. V., Hernandez, E. T., Ukraintsev, D. V., Guduru, R., Whiteley, M., and Anslyn, E. V. (2017) Dynamic covalent chemistry enables formation of antimicrobial peptide quaternary assemblies in a completely abiotic manner. Nat. Chem. 10, 45– 50, DOI: 10.1038/nchem.2847Google ScholarThere is no corresponding record for this reference.
- 19Diehl, K. L., Kolesnichenko, I. V., Robotham, S. A., Bachman, J. L., Zhong, Y., Brodbelt, J. S., and Anslyn, E. V. (2016) Click and chemically triggered declick reactions through reversible amine and thiol coupling via a conjugate acceptor. Nat. Chem. 8, 968– 973, DOI: 10.1038/nchem.2601Google Scholar19Click and chemically triggered declick reactions through reversible amine and thiol coupling via a conjugate acceptorDiehl, Katharine L.; Kolesnichenko, Igor V.; Robotham, Scott A.; Bachman, J. Logan; Zhong, Ye; Brodbelt, Jennifer S.; Anslyn, Eric V.Nature Chemistry (2016), 8 (10), 968-973CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The coupling and decoupling of mol. units is a fundamental undertaking of org. chem. Herein we report the use of a very simple conjugate acceptor, derived from Meldrum's acid, for the sequential 'clicking' together of an amine and a thiol in aq. conditions at neutral pH. Subsequently, this linkage can be 'declicked' by a chem. trigger to release the original amine and thiol undisturbed. The reactivity differs from that of other crosslinking agents because the selectivity for sequential functionalization derives from an altering of the electrophilicity of the conjugate acceptor on the addn. of the amine. We describe the use of the procedure to modify proteins, create multicomponent libraries and synthesize oligomers, all of which can be declicked to their starting components in a controlled fashion when desired. Owing to the mild reaction conditions and ease of use in a variety of applications, the method is predicted to have wide utility.
- 20Wang, T., Ng, D. Y. W., Wu, Y., Thomas, J., TamTran, T., and Weil, T. (2014) Bis-sulfide bioconjugates for glutathione triggered tumor responsive drug release. Chem. Commun. 50, 1116– 1118, DOI: 10.1039/C3CC47003BGoogle Scholar20Bis-sulfide bioconjugates for glutathione triggered tumor responsive drug releaseWang, Tao; Ng, David Y. W.; Wu, Yuzhou; Thomas, Jessica; TamTran, Thuy; Weil, TanjaChemical Communications (Cambridge, United Kingdom) (2014), 50 (9), 1116-1118CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)The reaction of bis-sulfone conjugation reagents with disulfide bonds allows the site-specific modification of various peptides and proteins. Herein, we present the intracellular disintegration of bis-sulfide contg. somatostatin bioconjugates under controlled, tumor-relevant glutathione levels. GSH responsive release is demonstrated, which offers high potential for designing tumor responsive therapeutics.
- 21Brustad, E., Bushey, M. L., Lee, J. W., Groff, D., Liu, W., and Schultz, P. G. (2008) A Genetically Encoded Boronate-Containing Amino Acid. Angew. Chem., Int. Ed. 47, 8220– 8223, DOI: 10.1002/anie.200803240Google Scholar21A genetically encoded boronate-containing amino acidBrustad, Eric; Bushey, Mark L.; Lee, Jae Wook; Groff, Dan; Liu, Wenshe; Schultz, Peter G.Angewandte Chemie, International Edition (2008), 47 (43), 8220-8223CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A biol. boronate: An orthogonal tRNA/aminoacyl-tRNA synthetase pair has been evolved for the genetic incorporation of a boronic acid into proteins. This amino acid has been used to purify proteins in a one-step scarless purifn. procedure as well as for the site-specific labeling of proteins using various boronic acid chemistries.
- 22Arzt, M., Seidler, C., Ng, D. Y. W., and Weil, T. (2014) Reversible Click Reactions with Boronic Acids to Build Supramolecular Architectures in Water. Chem. - Asian J. 9, 1994, DOI: 10.1002/asia.201402061Google Scholar22Reversible Click Reactions with Boronic Acids to Build Supramolecular Architectures in WaterArzt, Matthias; Seidler, Christiane; Ng, David Y. W.; Weil, TanjaChemistry - An Asian Journal (2014), 9 (8), 1994-2003CODEN: CAAJBI; ISSN:1861-4728. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The interaction of boronic acids with various bifunctional reagents offers great potential for the prepn. of responsive supramol. architectures. Boronic acids react with 1,2-diols yielding cyclic boronate esters that are stable at pH>7.4 but can be hydrolyzed at pH<5.0. The phenylboronic acid (PBA)-salicylhydroxamic acid (SHA) system offers ultra-fast reaction kinetics and high binding affinities. This Focus review summarizes the current advances in exploiting the bioorthogonal interaction of boronic acids to build pH-responsive supramol. architectures in water.
- 23Jackson, T. R., Springall, J. S., Rogalle, D., Masumoto, N., Ching Li, H., D’Hooge, F., Perera, S. P., Jenkins, T. A., James, T. D., Fossey, J. S., and van den Elsen, J. M. H. (2008) Boronate affinity saccharide electrophoresis: A novel carbohydrate analysis tool. Electrophoresis 29, 4185– 4191, DOI: 10.1002/elps.200800178Google ScholarThere is no corresponding record for this reference.
- 24Ng, D. Y. W., Arzt, M., Wu, Y., Kuan, S. L., Lamla, M., and Weil, T. (2014) Constructing Hybrid Protein Zymogens through Protective Dendritic Assembly. Angew. Chem., Int. Ed. 53, 324– 328, DOI: 10.1002/anie.201308533Google Scholar24Constructing Hybrid Protein Zymogens through Protective Dendritic AssemblyNg, David Y. W.; Arzt, Matthias; Wu, Yuzhou; Kuan, Seah Ling; Lamla, Markus; Weil, TanjaAngewandte Chemie, International Edition (2014), 53 (1), 324-328CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The modulation of protein uptake and activity in response to physiol. changes forms an integral part of smart protein therapeutics. We describe herein the self-assembly of a pH-responsive dendrimer shell onto the surface of active enzymes (trypsin, papain, DNase I) as a supramol. protecting group to form a hybrid dendrimer-enzyme complex. The attachment is based on the interaction between boronic acid and salicyl hydroxamate, thus allowing the macromol. assembly to respond to changes in pH between 5.0 and 7.4 in a highly reversible fashion. Catalytic activity is efficiently blocked in the presence of the dendrimer shell but is quant. restored upon shell degrdn. under acidic conditions. Unlike the native proteases, the hybrid constructs are shown to be efficiently taken up by A549 cells and colocalized in the acidic compartments. The programmed intracellular release of the proteases induced cytotoxicity, thereby uncovering a new avenue for precision biotherapeutics.
- 25Seidler, C., Ng, D. Y. W., Wu, Y., and Weil, T. (2016) pH responsive supramolecular core-shell protein hybrids. Supramol. Chem. 28, 742– 746, DOI: 10.1080/10610278.2016.1140764Google Scholar25pH responsive supramolecular core-shell protein hybridsSeidler, Christiane; Ng, David Y. W.; Wu, Yuzhou; Weil, TanjaSupramolecular Chemistry (2016), 28 (9-10), 742-746CODEN: SCHEER; ISSN:1029-0478. (Taylor & Francis Ltd.)PEGylation of proteins remains an integral part of macromol. therapeutics due to its well-known benign effects and pharmacokinetic enhancement properties. We report herein that PEGylation can be taken to the next level of complexity and dynamic behavior by introducing highly stable but responsive supramol. handles. By attaching small boronic acid groups onto proteins and salicylhydroxamate moiety to end-functionalise PEG chains, we demonstrate a comprehensive study on the facile assembly/disassembly of a core-shell protein-polymer architecture using fluorescence and microscale thermophoresis on a macromol. level. In addn., we demonstrate that both the activity and cellular transfer of functional proteins remained conserved throughout the assembly process thus establishing a rapid and orthogonal strategy towards protein PEGylation.
- 26Swaminathan, R., Ravi, V. K., Kumar, S., Kumar, M. V. S., and Chandra, N. (2011) Lysozyme: A model protein for amyloid research. Adv. Protein Chem. Struct. Biol. 84, 63– 111, DOI: 10.1016/B978-0-12-386483-3.00003-3Google Scholar26Lysozyme: A model protein for amyloid researchSwaminathan, Rajaram; Ravi, Vijay Kumar; Kumar, Satish; Kumar, Mattaparthi Venkata Satish; Chandra, NividhAdvances in Protein Chemistry and Structural Biology (2011), 84 (), 63-111CODEN: APCSG7; ISSN:1876-1623. (Elsevier Ltd.)A review. Ever since lysozyme was discovered by Fleming in 1922, this protein has emerged as a model for investigations on protein structure and function. Over the years, several high-resoln. structures have yielded a wealth of structural data on this protein. Extensive studies on folding of lysozyme have shown how different regions of this protein dynamically interact with one another. Data is also available from numerous biotechnol. studies wherein lysozyme has been employed as a model protein for recovering active recombinant protein from inclusion bodies using small mols. like L-arginine. A variety of conditions have been developed in vitro to induce fibrillation in hen lysozyme. They include (a) acidic pH at elevated temp., (b) concd. solns. of ethanol, (c) moderate concns. of guanidinium hydrochloride at moderate temp., and (d) alk. pH at room temp. This review aims to bring together similarities and differences in aggregation mechanisms, morphol. of aggregates, and related issues that arise using the different conditions mentioned above to improve our understanding. The alk. pH condition (pH 12.2), discovered and studied extensively in our lab, shall receive special attention. More than a decade ago, it was revealed that mutations in human lysozyme can cause accumulation of large quantities of amyloid in liver, kidney, and other regions of gastrointestinal tract. Understanding the mechanism of lysozyme aggregation will probably have therapeutic implications for the treatment of systemic nonneuropathic amyloidosis. Numerous studies have begun to focus attention on inhibition of lysozyme aggregation using antibody or small mols. The enzymic activity of lysozyme presents a convenient handle to quantify the native population of lysozyme in a sample where aggregation has been inhibited. The rich information available on lysozyme coupled with the multiple conditions that have been successful in inducing/inhibiting its aggregation in vitro makes lysozyme an ideal model protein to investigate amyloidogenesis.
- 27Rambaran, R. N. and Serpell, L. C. (2008) Amyloid fibrils: abnormal protein assembly. Prion 2, 112– 7, DOI: 10.4161/pri.2.3.7488Google Scholar27Amyloid fibrils: abnormal protein assemblyRambaran Roma N; Serpell Louise CPrion (2008), 2 (3), 112-7 ISSN:.Amyloid refers to the abnormal fibrous, extracellular, proteinaceous deposits found in organs and tissues. Amyloid is insoluble and is structurally dominated by beta-sheet structure. Unlike other fibrous proteins it does not commonly have a structural, supportive or motility role but is associated with the pathology seen in a range of diseases known as the amyloidoses. These diseases include Alzheimer's, the spongiform encephalopathies and type II diabetes, all of which are progressive disorders with associated high morbidity and mortality. Not surprisingly, research into the physicochemical properties of amyloid and its formation is currently intensely pursued. In this chapter we will highlight the key scientific findings and discuss how the stability of amyloid fibrils impacts on bionanotechnology.
- 28Mankar, S., Anoop, A., Sen, S., and Maji, S. K. (2011) Nanomaterials: amyloids reflect their brighter side. Nano Rev. 2, 6032, DOI: 10.3402/nano.v2i0.6032Google ScholarThere is no corresponding record for this reference.
- 29Meier, C. and Welland, M. E. (2011) Wet-Spinning of Amyloid Protein Nanofibers into Multifunctional High-Performance Biofibers. Biomacromolecules 12, 3453– 3459, DOI: 10.1021/bm2005752Google Scholar29Wet-Spinning of Amyloid Protein Nanofibers into Multifunctional High-Performance BiofibersMeier, Christoph; Welland, Mark E.Biomacromolecules (2011), 12 (10), 3453-3459CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Amyloid nanofibers derived from hen egg white lysozyme were processed into macroscopic fibers in a wet-spinning process based on interfacial polyion complexation using a polyanionic polysaccharide as crosslinker. As a result of their amyloid nanostructure, the hierarchically self-assembled protein fibers have a stiffness of up to 14 GPa and a tensile strength of up to 326 MPa. Fine-tuning of the polyelectrolytic interactions via pH allows to trigger the release of small mols., as demonstrated with riboflavin-5'-phosphate. The amyloid fibrils, highly oriented within the gellan gum matrix, were mineralized with calcium phosphate, mimicking the fibrolamellar structure of bone. The formed mineral crystals are highly oriented along the nanofibers, thus resulting in a 9-fold increase in fiber stiffness.
- 30Wang, T., Riegger, A., Lamla, M., Wiese, S., Oeckl, P., Otto, M., Wu, Y., Fischer, S., Barth, H., Kuan, S. L., and Weil, T. (2016) Water-soluble allyl sulfones for dual site-specific labelling of proteins and cyclic peptides. Chem. Sci. 7, 3234– 3239, DOI: 10.1039/C6SC00005CGoogle Scholar30Water-soluble allyl sulfones for dual site-specific labelling of proteins and cyclic peptidesWang, Tao; Riegger, Andreas; Lamla, Markus; Wiese, Sebastian; Oeckl, Patrick; Otto, Markus; Wu, Yuzhou; Fischer, Stephan; Barth, Holger; Kuan, Seah Ling; Weil, TanjaChemical Science (2016), 7 (5), 3234-3239CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Water-sol. allyl sulfones provide convenient site-specific disulfide rebridging of native proteins and cyclic peptides. The site-selective functionalization of (a) the peptide hormone somatostatin, (b) the interchain disulfide of bovine insulin and (c) functionalization of the proteins GFP and lysozyme with allyl sulfones proceeds in aq. soln. Allyl sulfones offer three functionalizable sites that react with thiol contg. mols. in a step-wise fashion. Dual labeling of proteins and cyclic peptides is achieved attachment of chromophore and affinity tag in a single reaction step, which is of great significance for the construction of precise multifunctional peptide and protein conjugates.
- 31Brocchini, S., Balan, S., Godwin, A., Choi, J.-W., Zloh, M., and Shaunak, S. (2006) PEGylation of native disulfide bonds in proteins. Nat. Chem. Biol. 1, 2241– 2252, DOI: 10.1038/nprot.2006.346Google ScholarThere is no corresponding record for this reference.
- 32Robinson, E., Nunes, J. P. M., Vassileva, V., Maruani, A., Nogueira, J. C. F., Smith, M. E. B., Pedley, R. B., Caddick, S., Baker, J. R., and Chudasama, V. (2017) Pyridazinediones deliver potent, stable, targeted and efficacious antibody–drug conjugates (ADCs) with a controlled loading of 4 drugs per antibody. RSC Adv. 7, 9073– 9077, DOI: 10.1039/C7RA00788DGoogle Scholar32Pyridazinediones deliver potent, stable, targeted and efficacious antibody-drug conjugates (ADCs) with a controlled loading of 4 drugs per antibodyRobinson, Eifion; Nunes, Joao P. M.; Vassileva, Vessela; Maruani, Antoine; Nogueira, Joao C. F.; Smith, Mark E. B.; Pedley, R. Barbara; Caddick, Stephen; Baker, James R.; Chudasama, VijayRSC Advances (2017), 7 (15), 9073-9077CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)Herein we report the use of pyridazinediones to functionalize the native solvent accessible interstrand disulfide bonds in trastuzumab with monomethyl auristatin E (MMAE). This method of conjugation delivers serum stable antibody-drug conjugates (ADCs) with a controlled drug loading of 4. Moreover, we demonstrate that the MMAE-bearing ADCs are potent, selective and efficacious against cancer cell lines in both in vitro and in vivo models.
- 33Greene, M. K., Richards, D. A., Nogueira, J. C. F., Campbell, K., Smyth, P., Fernández, M., Scott, C. J., and Chudasama, V. (2018) Forming next-generation antibody–nanoparticle conjugates through the oriented installation of non-engineered antibody fragments. Chem. Sci. 9, 79– 87, DOI: 10.1039/C7SC02747HGoogle Scholar33Forming next-generation antibody-nanoparticle conjugates through the oriented installation of non-engineered antibody fragmentsGreene, Michelle K.; Richards, Daniel A.; Nogueira, Joao C. F.; Campbell, Katrina; Smyth, Peter; Fernandez, Marcos; Scott, Christopher J.; Chudasama, VijayChemical Science (2018), 9 (1), 79-87CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The successful development of targeted nanotherapeutics is contingent upon the conjugation of therapeutic nanoparticles to target-specific ligands, with particular emphasis being placed on antibody-based ligands. Thus, new methods that enable the covalent and precise installation of targeting antibodies to nanoparticle surfaces are greatly desired, esp. those which do not rely on costly and time-consuming antibody engineering techniques. Herein we present a novel method for the highly controlled and oriented covalent conjugation of non-engineered antibody F(ab) fragments to PLGA-PEG nanoparticles using disulfide-selective pyridazinedione linkers and strain-promoted alkyne-azide click chem. Exemplification of this method with trastuzumab and cetuximab showed significant improvements in both conjugation efficiency and antigen binding capability, when compared to commonly employed strategies for antibody-nanoparticle construction. This new approach paves the way for the development of antibody-targeted nanomedicines with improved paratope availability, reproducibility and uniformity to enhance both biol. activity and ease of manuf.
- 34Trivedi, M. V., Laurence, J. S., and Siahaan, T. J. (2009) The role of thiols and disulfides on protein stability. Curr. Protein Pept. Sci. 10, 614– 25, DOI: 10.2174/138920309789630534Google Scholar34The role of thiols and disulfides on protein stabilityTrivedi, Maulik V.; Laurence, Jennifer S.; Siahaan, Teruna J.Current Protein and Peptide Science (2009), 10 (6), 614-625CODEN: CPPSCM; ISSN:1389-2037. (Bentham Science Publishers Ltd.)A review. There has been a tremendous increase in the no. of approved drugs derived from recombinant proteins; however, their development as potential drugs has been hampered by their instability that causes difficulty to formulate them as therapeutic agents. It has been shown that the reactivity of thiol and disulfide functional groups could catalyze chem. (i.e., oxidn. and β-elimination reactions) and phys. (i.e., aggregation and pptn.) degrdn. of proteins. Because most proteins contain a free Cys residue and/or a disulfide bond, this review is focused on their roles in the phys. and chem. stability of proteins. The effect of introducing a disulfide bond to improve phys. stability of proteins and the mechanisms of degrdn. of disulfide bond were discussed. The qual./quant. methods to det. the presence of the thiol in the Cys residue and various methods to derivatize the thiol group for improving protein stability were also illustrated.
- 35Rosen, C. B. and Francis, M. B. (2017) Targeting the N terminus for site-selective protein modification. Nat. Chem. Biol. 13, 697– 705, DOI: 10.1038/nchembio.2416Google Scholar35Targeting the N terminus for site-selective protein modificationRosen, Christian B.; Francis, Matthew B.Nature Chemical Biology (2017), 13 (7), 697-705CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. The formation of well-defined protein bioconjugates is crit. for many studies and technologies in chem. biol. Tried-and-true methods for accomplishing this typically involve the targeting of cysteine residues, but the rapid growth of contemporary bioconjugate applications has required an expanded repertoire of modification techniques. One very powerful set of strategies involves the modification of proteins at their N termini, as these positions are typically solvent exposed and provide chem. distinct sites for many protein targets. Several chem. techniques can be used to modify N-terminal amino acids directly or convert them into unique functional groups for further ligations. A growing no. of N-terminus-specific enzymic ligation strategies provided addnl. possibilities. This Perspective provides an overview of N-terminal modification techniques and the chem. rationale governing each. Examples of specific N-terminal protein conjugates are provided, along with their uses in a no. of diverse biol. applications.
- 36Tanaka, K., Fukase, K., and Katsumura, S. (2011) Exploring a Unique Reactivity of 6p-Azaelectrocyclization to Enzyme Inhibition, Natural Products Synthesis, and Molecular Imaging: An Approach to Chemical Biology by Synthetic Chemists. Synlett 2011, 2115– 2139, DOI: 10.1055/s-0030-1261192Google ScholarThere is no corresponding record for this reference.
- 37Ban, H., Nagano, M., Gavrilyuk, J., Hakamata, W., Inokuma, T., and Barbas, C. F. (2013) Facile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click Reaction. Bioconjugate Chem. 24, 520– 532, DOI: 10.1021/bc300665tGoogle Scholar37Facile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click ReactionBan, Hitoshi; Nagano, Masanobu; Gavrilyuk, Julia; Hakamata, Wataru; Inokuma, Tsubasa; Barbas, Carlos F., IIIBioconjugate Chemistry (2013), 24 (4), 520-532CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The scope, chemoselectivity, and utility of the click-like tyrosine labeling reaction with 4-phenyl-3H-1,2,4-triazoline-3,5(4H)-diones (PTADs) is reported. To study the utility and chemoselectivity of PTAD derivs. in peptide and protein chem., we synthesized PTAD derivs. possessing azide, alkyne, and ketone groups and studied their reactions with amino acid derivs. and peptides of increasing complexity. With proteins we studied the compatibility of the tyrosine click reaction with cysteine and lysine-targeted labeling approaches and demonstrate that chemoselective trifunctionalization of proteins is readily achieved. In particular cases, we noted that PTAD decompn. resulted in formation of a putative isocyanate byproduct that was promiscuous in labeling. This side reaction product, however, was readily scavenged by the addn. of a small amt. of 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) to the reaction medium. To study the potential of the tyrosine click reaction to introduce poly(ethylene glycol) chains onto proteins (PEGylation), we demonstrate that this novel reagent provides for the selective PEGylation of chymotrypsinogen, whereas traditional succinimide-based PEGylation targeting lysine residues provided a more diverse range of PEGylated products. Finally, we applied the tyrosine click reaction to create a novel antibody-drug conjugate. For this purpose, we synthesized a PTAD deriv. linked to the HIV entry inhibitor aplaviroc. Labeling of the antibody trastuzumab with this reagent provided a labeled antibody conjugate that demonstrated potent HIV-1 neutralization activity demonstrating the potential of this reaction in creating protein conjugates with small mols. The tyrosine click linkage demonstrated stability to extremes of pH, temp., and exposure to human blood plasma indicating that this linkage is significantly more robust than maleimide-type linkages that are commonly employed in bioconjugations. These studies support the broad utility of this reaction in the chemoselective modification of small mols., peptides, and proteins under mild aq. conditions over a broad pH range using a wide variety of biol. acceptable buffers such as phosphate buffered saline (PBS) and 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffers as well as others and mixed buffered compns.
- 38Santos, F. M. F., Rosa, J. N., Candeias, N. R., Carvalho, C. P., Matos, A. I., Ventura, A. E., Florindo, H. F., Silva, L. C., Pischel, U., and Gois, P. M. P. (2016) A Three-Component Assembly Promoted by Boronic Acids Delivers a Modular Fluorophore Platform (BASHY Dyes). Chem. - Eur. J. 22, 1631– 1637, DOI: 10.1002/chem.201503943Google Scholar38A Three-Component Assembly Promoted by Boronic Acids Delivers a Modular Fluorophore Platform (BASHY Dyes)Santos, Fabio M. F.; Rosa, Joao N.; Candeias, Nuno R.; Carvalho, Catia Parente; Matos, Ana I.; Ventura, Ana E.; Florindo, Helena F.; Silva, Liana C.; Pischel, Uwe; Gois, Pedro M. P.Chemistry - A European Journal (2016), 22 (5), 1631-1637CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)The modular assembly of boronic acids with Schiff-base ligands enabled the construction of innovative fluorescent dyes [boronic acid salicylidenehydrazone (BASHY)] with suitable structural and photophys. properties for live cell bioimaging applications. This reaction enabled the straightforward synthesis (yields up to 99 %) of structurally diverse and photostable dyes that exhibit a polarity-sensitive green-to-yellow emission with high quantum yields of up to 0.6 in nonpolar environments. These dyes displayed a high brightness (up to 54 000 M-1 cm-1). The promising structural and fluorescence properties of BASHY dyes fostered the prepn. of non-cytotoxic, stable, and highly fluorescent poly(lactide-co-glycolide) nanoparticles that were effectively internalized by dendritic cells. The dyes were also shown to selectively stain lipid droplets in HeLa cells, without inducing any appreciable cytotoxicity or competing plasma membrane labeling; this confirmed their potential as fluorescent stains.
- 39Cal, P. M. S. D., Sieglitz, F., Santos, F. M. F., Parente Carvalho, C., Guerreiro, A., Bertoldo, J. B., Pischel, U., Gois, P. M. P., and Bernardes, G. J. L. (2017) Site-selective installation of BASHY fluorescent dyes to Annexin V for targeted detection of apoptotic cells. Chem. Commun. 53, 368– 371, DOI: 10.1039/C6CC08671CGoogle Scholar39Site-selective installation of BASHY fluorescent dyes to Annexin V for targeted detection of apoptotic cellsCal, Pedro M. S. D.; Sieglitz, Florian; Santos, Fabio M. F.; Parente Carvalho, Catia; Guerreiro, Ana; Bertoldo, Jean B.; Pischel, Uwe; Gois, Pedro M. P.; Bernardes, Goncalo J. L.Chemical Communications (Cambridge, United Kingdom) (2017), 53 (2), 368-371CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Fluorophores are indispensable for imaging biol. processes. The authors report the design and synthesis of azide-tagged boronic acid salicylidenehydrazone (BASHY) dyes and their use for site-selective labeling of Annexin V. The Annexin V-BASHY conjugate maintained function and fluorescence as demonstrated by the targeted detection of apoptotic cells.
- 40Ng, D. Y. W., Vill, R., Wu, Y., Koynov, K., Tokura, Y., Liu, W., Sihler, S., Kreyes, A., Ritz, S., Barth, H., Ziener, U., and Weil, T. (2017) Directing intracellular supramolecular assembly with N-heteroaromatic quaterthiophene analogues. Nat. Commun. 8, 1850, DOI: 10.1038/s41467-017-02020-2Google Scholar40Directing intracellular supramolecular assembly with N-heteroaromatic quaterthiophene analoguesNg David Y W; Wu Yuzhou; Koynov Kaloian; Tokura Yu; Liu Weina; Weil Tanja; Vill Roman; Tokura Yu; Liu Weina; Sihler Susanne; Kreyes Andreas; Ziener Ulrich; Weil Tanja; Ritz Sandra; Barth HolgerNature communications (2017), 8 (1), 1850 ISSN:.Self-assembly in situ, where synthetic molecules are programmed to organize in a specific and complex environment i.e., within living cells, can be a unique strategy to influence cellular functions. Here we present a small series of rationally designed oligothiophene analogues that specifically target, locate and dynamically self-report their supramolecular behavior within the confinement of a cell. Through the recognition of the terminal alkyl substituent and the amphiphilic pyridine motif, we show that the cell provides different complementary pathways for self-assembly that can be traced easily with fluorescence microscopy as their molecular organization emits in distinct fluorescent bands. Importantly, the control and induction of both forms are achieved by time, temperature and the use of the intracellular transport inhibitor, bafilomycin A1. We showcase the importance of both intrinsic (cell) and extrinsic (stimulus) factors for self-organization and the potential of such a platform toward developing synthetic functional components within living cells.
- 41Najera, C., Baldo, B., and Yus, M. (1988) Regio- and Stereo-selective Synthesis of β-Sulphonyl-a,β- Unsaturated Carbonyl Compounds. J. Chem. Soc., Perkin Trans. 1 1029, DOI: 10.1039/P19880001029Google ScholarThere is no corresponding record for this reference.
- 42Guez, V., Roux, P., Navon, A., and Goldberg, M. E. (2002) Role of individual disulfide bonds in hen lysozyme early folding steps. Protein Sci. 11, 1136– 51, DOI: 10.1110/ps.3960102Google Scholar42Role of individual disulfide bonds in hen lysozyme early folding stepsGuez, Valerie; Roux, Pascale; Navon, Amiel; Goldberg, Michel E.Protein Science (2002), 11 (5), 1136-1151CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)To probe the role of individual disulfide bonds in the folding kinetics of hen lysozyme, variants with 2 mutations, C30A/C115A, C64A/C80A, and C76A/C94A, were constructed. The corresponding proteins, each lacking one disulfide bond, were produced in Escherichia coli as inclusion bodies and solubilized, purified, and renatured/oxidized using original protocols. Their enzymic, spectral, and hydrodynamic characteristics confirmed that their conformations were very similar to that of native wild-type (WT) lysozyme. Stopped-flow studies on the renaturation of these guanidine-HCl-unfolded proteins with their 3 disulfide bonds intact showed that, for the 3 variants, the native far-UV ellipticity was regained in a burst phase within the 4-ms instrument dead-time. The transient overshoots of far-UV ellipticity and Trp residue fluorescence that follow the burst phase, as well as the kinetics of transient 8-anilino-1-naphthalene-sulfonic acid (ANS) binding, were diversely affected depending on the variant. Together with previous reports on the folding kinetics of WT lysozyme carboxymethylated on Cys-6 and Cys-127, detailed anal. of the kinetics showed that: (1) none of the disulfide bonds were indispensable for the rapid formation (<4 ms) of the native-like secondary structure; (2) the 2 intra-α-domain disulfides (Cys-6-Cys-127 and Cys-30-Cys-115) must be simultaneously present to generate the trapped intermediate responsible for the slow folding population obsd. in WT lysozyme; and (3) the intra-β-domain (Cys-64-Cys-80) and the inter-αβ-domains (Cys-76-Cys-94) disulfide bonds did not affect the kinetics of formation of the trapped intermediate but were involved in its stability.
- 43Springsteen, G. and Wang, B. (2002) A detailed examination of boronic acid–diol complexation. Tetrahedron 58, 5291– 5300, DOI: 10.1016/S0040-4020(02)00489-1Google Scholar43A detailed examination of boronic acid-diol complexationSpringsteen, Greg; Wang, BingheTetrahedron (2002), 58 (26), 5291-5300CODEN: TETRAB; ISSN:0040-4020. (Elsevier Science Ltd.)Boronic acids bind with compds. contg. diol moieties with high affinity through reversible boronate formation. However, the conditions that foster tight binding between the diol and the boronic acid are not well understood. Also, due to the multiple ionic states of both the boronic acid and boronate ester, the equil. consts. reported in the literature have not always been strictly defined, and therefore there is a lack of comparability between the reported values. To address these issues, a method was developed for examg. boronate ester stability using the fluorescent reporter Alizarin Red S. and this system has been used to det. the binding consts. of a series of diols, and as a basis from which to derive a no. of relationships that correlate the various equil. consts. in the literature.
- 44Shugar, D. (1952) The measurement of lysozyme activity and the ultra-violet inactivation of lysozyme. Biochim. Biophys. Acta 8, 302– 9, DOI: 10.1016/0006-3002(52)90045-0Google Scholar44Measurement of lysozyme activity and the ultraviolet inactivation of lysozymeShugar, DavidBiochimica et Biophysica Acta (1952), 8 (), 302-9CODEN: BBACAQ; ISSN:0006-3002.Lysozyme (I) activity is detd. by measuring the decrease with time of the optical d. of a suspension of Micrococcus lysodeikticus. The analysis is made at pH 7.1 (M/15 phosphate buffer) in 3 ml. vol. (optical d. 0.5 to 0.75) to which 0.02-0.05 cc. of enzyme soln. is added. The readings are made at 2820 A. over a 4.5 min. period. Inactivation by ultraviolet light is a first order reaction, k = 0.158 min.-1. The quantum yield for inactivation at 2537 A. is 0.024 over the pH range 3.6-12.0. No apparent liberation of peptides or amino acids was observed but some photo-oxidation took place.
- 45Davies, R. C. and Neuberger, A. (1969) Modification of lysine and arginine residues of lysozyme and the effect of enzymatic activity. Biochim. Biophys. Acta - Enzymol. 178, 306– 317, DOI: 10.1016/0005-2744(69)90398-2Google Scholar45Modification of lysine and arginine residues of lysozyme and the effect on enzymic activityDavies, Richard C.; Neuberger, AlbertBiochimica et Biophysica Acta, Enzymology (1969), 178 (2), 306-17CODEN: BBEZAD; ISSN:0924-1086.Acetylation of all 6 lysine residues of lysozyme (EC 3.2.1.17) abolished lytic action towards cells of Micrococcus lysodeikticus in 0.05M phosphate buffer (pH 6.2) but did not affect cleavage of the tetramer of N-acetylglucosamine (GlcNAc)4 obtained from chitin. Acetylated lysozyme still lysed cells in solns. of low ionic strength at pH 6.2. Compared with unmodified enzyme the activity profile for acetyl lysozyme was displaced to much lower values of ionic strength and was still markedly dependent on pH. Chem. modification of the lysine groups with ethyl acetamidate increased the activity towards cells slightly, yet did not alter the activity towards (GlcNAc)4. Modification of 7 out of 8 arginine residues with 2,3-butanedione in borate buffer reduced the activity of acetyl lysozyme towards cells but not towards (GlcNAc)4. Since all the basic residues of lysozyme apparently lie outside the active center, the persistence of lytic activity over a very wide pH range is discussed in terms of the currently accepted mechanism of action of lysozyme.
- 46Imoto, T., Moriyama, S., and Yagishita, K. (1976) A Study of the Native-Denatured (N to/from D) Transition in Lysozyme III. Effect of Alteration of Net Charge by Acetylation. J. Biochem. 80, 1319– 1325, DOI: 10.1093/oxfordjournals.jbchem.a131404Google Scholar46A study of the native-denatured (N .dblharw. D) transition in lysozyme. III. Effect of alteration of net charge by acetylationImoto, Taiji; Moriyama, Sigeru; Yagishita, KazuyoshiJournal of Biochemistry (1976), 80 (6), 1319-25CODEN: JOBIAO; ISSN:0021-924X.Measurement of the enzymic activity and fluorescence properties showed that the gross conformation of acetylated lysozyme (I) is very similar to that of native I. On the other hand, protease digestion, t-Bu hypochloride modification, and thermal denaturation of native, acetylated, and guanidinated I showed that acetylation causes a small but significant shift of the N.dblharw.D transition to the right. Thus, charge balance in a protein plays an important role in maintaining its conformation. The difference between equil. and kinetic methods of monitoring protein denaturation was also clarified.
- 47Masuda, T., Kitabatake, N., and Ide, N. (2005) Effects of Chemical Modification of Lysine Residues on the Sweetness of Lysozyme. Chem. Chem. Senses 30, 253– 264, DOI: 10.1093/chemse/bji021Google Scholar47Effects of Chemical Modification of Lysine Residues on the Sweetness of LysozymeMasuda, Tetsuya; Ide, Nobuyuki; Kitabatake, NaofumiChemical Senses (2005), 30 (3), 253-264CODEN: CHSED8; ISSN:0379-864X. (Oxford University Press)Lysozyme is a sweet-tasting protein with a sweetness threshold value of around 7 μM. To clarify the effect of basicity at the side chain of lysine residues on the threshold values of sweetness, charge-specific chem. modifications such as guanidination, acetylation and phosphopyridoxylation of lysine residues were performed. Sensory anal. showed that the sweetness threshold value of lysozyme was not changed by guanidination, whereas it was increased markedly by acetylation and phosphopyridoxylation. To confirm the importance of the basicity in the lysine residues in detail, purifn. of acetylated (Ac-) and phosphopyridoxylated (PLP-) lysozymes using SP-ion exchange column chromatog. was performed. The threshold values were not changed by modification with fewer than two residues (∼7 μM), whereas the threshold values significantly increased to 15 and 34 μM when tetra-Ac and tri-PLP, resp. Furthermore, sweetness was not detected at 30 μM (hexa-, penta-Ac and tetra-PLP). It should be noted that removal of the neg. charges of the phosphate groups in the tri-PLP lysozyme by acid phosphatase resulted in the recovery of sweetness (6.4 μM), indicating that basicity at the position of the lysine residues is responsible for lysozyme sweetness and that strict charge complementarities might be required for interaction to its putative receptor.
- 48O’Brien, E. C., Farkas, E., Gil, M. J., Fitzgerald, D., Castineras, A., and Nolan, K. B. (2000) Metal complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid and benzohydroxamic acid. Crystal and molecular structure of [Cu(phen)2(Cl)]Cl·H2Sha, a model for a peroxidase-inhibitor complex. J. Inorg. Biochem. 79, 47– 51, DOI: 10.1016/S0162-0134(99)00245-7Google Scholar48Metal complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid and benzohydroxamic acid. Crystal and molecular structure of [Cu(phen)2(Cl)]Cl·H2Sha, a model for a peroxidase-inhibitor complexO'Brien, Eimear C.; Farkas, Etelka; Gil, Marie Jose; Fitzgerald, Desmond; Castineras, Alfonso; Nolan, Kevin B.Journal of Inorganic Biochemistry (2000), 79 (1-4), 47-51CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier Science Inc.)Stability consts. of iron(III), copper(II), nickel(II) and zinc(II) complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid (HAha) and benzohydroxamic acid (HBha) have been detd. at 25.0°C, I=0.2 mol dm-3 KCl in aq. soln. The complex stability order, iron(III) » copper(II) > nickel(II) ∼ zinc(II) was obsd. while complexes of H2Sha were found to be more stable than those of the other two ligands. In the prepn. of ternary metal ion complexes of these ligands and 1,10-phenanthroline (phen) the cryst. complex [Cu(phen)2(Cl)]Cl·H2Sha was obtained and its crystal structure detd. This complex is a model for hydroxamate-peroxidase inhibitor interactions.
- 49Yang, S.-M., Lagu, B., and Wilson, L. J. (2007) Mild and Efficient Lewis Acid-Promoted Detritylation in the Synthesis of N -Hydroxy Amides: A Concise Synthesis of (−)-Cobactin T. J. Org. Chem. 72, 8123– 8126, DOI: 10.1021/jo701411dGoogle ScholarThere is no corresponding record for this reference.
- 50Shin, S. B. Y., Almeida, R. D., Gerona-Navarro, G., Bracken, C., and Jaffrey, S. R. (2010) Assembling ligands in situ using bioorthogonal boronate ester synthesis. Chem. Biol. 17, 1171– 6, DOI: 10.1016/j.chembiol.2010.09.008Google Scholar50Assembling ligands in situ using bioorthogonal boronate ester synthesisShin, Sung Bin Y.; Almeida, Ramiro D.; Gerona-Navarro, Guillermo; Bracken, Clay; Jaffrey, Samie R.Chemistry & Biology (Cambridge, MA, United States) (2010), 17 (11), 1171-1176CODEN: CBOLE2; ISSN:1074-5521. (Cell Press)Many mols. that could manipulate cellular function are not practical due to their large size and concomitant undesirable pharmacokinetic properties. Here, we describe a bioorthogonal, highly stable boronate ester (HiSBE) synthesis and use this reaction to synthesize a biol. active mol. from smaller precursors in a physiol. context. The rapid rate of HiSBE synthesis suggests that it may be useful for assembling a wide variety of biol. active mols. in physiol. solns.
- 51Shrestha, D., Jenei, A., Nagy, P., Vereb, G., and Szöllősi, J. (2015) Understanding FRET as a research tool for cellular studies. Int. J. Mol. Sci. 16, 6718– 56, DOI: 10.3390/ijms16046718Google Scholar51Understanding FRET as a research tool for cellular studiesShrestha, Dilip; Jenei, Attila; Nagy, Peter; Vereb, Gyoergy; Szoellosi, JanosInternational Journal of Molecular Sciences (2015), 16 (4), 6718-6756CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)Communication of mol. species through dynamic assocn. and/or dissocn. at various cellular sites governs biol. functions. Understanding these physiol. processes require delineation of mol. events occurring at the level of individual complexes in a living cell. Among the few non-invasive approaches with nanometer resoln. are methods based on Forster Resonance Energy Transfer (FRET). FRET is effective at a distance of 1-10 nm which is equiv. to the size of macromols., thus providing an unprecedented level of detail on mol. interactions. The emergence of fluorescent proteins and SNAP- and CLIP- tag proteins provided FRET with the capability to monitor changes in a mol. complex in real-time making it possible to establish the functional significance of the studied mols. in a native environment. Now, FRET is widely used in biol. sciences, including the field of proteomics, signal transduction, diagnostics and drug development to address questions almost unimaginable with biochem. methods and conventional microscopies. However, the underlying physics of FRET often scares biologists. Therefore, in this review, our goal is to introduce FRET to non-physicists in a lucid manner. We will also discuss our contributions to various FRET methodologies based on microscopy and flow cytometry, while describing its application for detg. the mol. heterogeneity of the plasma membrane in various cell types.
- 52Yano, Y. and Matsuzaki, K. (2009) Tag–probe labeling methods for live-cell imaging of membrane proteins. Biochim. Biophys. Acta, Biomembr. 1788, 2124– 2131, DOI: 10.1016/j.bbamem.2009.07.017Google Scholar52Tag-probe labeling methods for live-cell imaging of membrane proteinsYano, Yoshiaki; Matsuzaki, KatsumiBiochimica et Biophysica Acta, Biomembranes (2009), 1788 (10), 2124-2131CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)A review. Instead of using reconstituted proteoliposomes, in situ investigations of membrane proteins in living cell membranes are important because the heterogeneous and dynamic nature of biomembranes significantly affects their behavior. Protein-specific labeling is a key technique for the detection of a target protein by fluorescence measurements, particularly fluorescence microscopy. However, conventional genetic fusion with fluorescent proteins has several shortcomings. Post-translational labeling methods using a genetically encodable tag and synthetic probes targeting to the tag can overcome these limitations. This review summarizes emerging tag-probe techniques for labeling specific membrane proteins and their applications, including endocytic internalization, partitioning to specific membrane domains, interprotein interactions, and conformational changes.
- 53Soriano-Ursúa, M. A., Farfán-García, E. D., López-Cabrera, Y., Querejeta, E., and Trujillo-Ferrara, J. G. (2014) Boron-containing acids: Preliminary evaluation of acute toxicity and access to the brain determined by Raman scattering spectroscopy. NeuroToxicology 40, 8– 15, DOI: 10.1016/j.neuro.2013.10.005Google Scholar53Boron-containing acids: Preliminary evaluation of acute toxicity and access to the brain determined by Raman scattering spectroscopySoriano-Ursua, Marvin A.; Farfan-Garcia, Eunice D.; Lopez-Cabrera, Yessica; Querejeta, Enrique; Trujillo-Ferrara, Jose G.NeuroToxicology (2014), 40 (), 8-15CODEN: NRTXDN; ISSN:0161-813X. (Elsevier Inc.)Boron-contg. compds. (BCCs), particularly boron contg. acids (BCAs), have become attractive moieties or mols. in drug development. It has been suggested that when functional groups with boron atoms are added to well-known drugs, the latter are conferred with greater potency and efficacy in relation to their target receptors. However, the use of BCAs in drug development is limited due to the lack of a toxicol. profile. Consequently, the aim of the present study was to evaluate the acute toxicity of boric and boronic acids. Thus, a detn. was made of the LD (LD50) of test compds. in male CD1 mice, as well as the ED required to neg. affect spontaneous motor activity and to produce notable behavioral abnormalities. After treatment of animals at different doses, macroscopic observations were made from a necropsy, and Raman scattering spectroscopic studies were carried out on brain tissue samples. In general, the results show that most of the tested BCAs have very low toxicity, evidenced by the high doses required to induce notable toxic effects (greater than 100 mg/kg of body wt. for all compds., except for 3-thyenilboronic acid). Such toxic effects, presumably mediated by action on the CNS, include eye damage, gastrointestinal effects (e.g., gastric-gut dilatation and fecal retention), sedation, hypnosis and/or trembling. This preliminary toxicol. profile suggests that BCAs can be considered potential therapeutic agents or moieties to be added to other compds. in the development of new drugs. Future studies are required to explore possible chronic toxicity of BCCs.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 41 publications.
- Ferran Esteve, Jean-Louis Schmitt, Sergii Kolodych, Oleksandr Koniev, Jean-Marie Lehn. Selective Protein (Post-)modifications through Dynamic Covalent Chemistry: Self-activated SNAr Reactions. Journal of the American Chemical Society 2025, 147
(2)
, 2049-2060. https://doi.org/10.1021/jacs.4c15421
- Mylène Richard, Soizic Martin Aubert, Caroline Denis, Steven Dubois, Hervé Nozach, Charles Truillet, Bertrand Kuhnast. Fluorine-18 and Radiometal Labeling of Biomolecules via Disulfide Rebridging. Bioconjugate Chemistry 2023, 34
(11)
, 2123-2132. https://doi.org/10.1021/acs.bioconjchem.3c00440
- Bente A. Somsen, Rick J.C. Schellekens, Carlo J.A. Verhoef, Michelle R. Arkin, Christian Ottmann, Peter J. Cossar, Luc Brunsveld. Reversible Dual-Covalent Molecular Locking of the 14-3-3/ERRγ Protein–Protein Interaction as a Molecular Glue Drug Discovery Approach. Journal of the American Chemical Society 2023, 145
(12)
, 6741-6752. https://doi.org/10.1021/jacs.2c12781
- Michael J. Swierczynski, Yuxuan Ding, Zachary T. Ball. Dual-Boronic Acid Reagents That Combine Dynamic and Covalent Bioconjugation. Bioconjugate Chemistry 2022, 33
(12)
, 2307-2313. https://doi.org/10.1021/acs.bioconjchem.2c00508
- Daniele Melodia, Zachary Di Pietro, Cheng Cao, Martina H. Stenzel, Robert Chapman. Traceless pH-Sensitive Antibody Conjugation Inspired by Citraconic Anhydride. Biomacromolecules 2022, 23
(12)
, 5322-5329. https://doi.org/10.1021/acs.biomac.2c01125
- Maksymilian Marek Zegota, Michael Andreas Müller, Bellinda Lantzberg, Gönül Kizilsavas, Jaime A. S. Coelho, Pierpaolo Moscariello, María Martínez-Negro, Svenja Morsbach, Pedro M. P. Gois, Manfred Wagner, David Y. W. Ng, Seah Ling Kuan, Tanja Weil. Dual Stimuli-Responsive Dynamic Covalent Peptide Tags: Toward Sequence-Controlled Release in Tumor-like Microenvironments. Journal of the American Chemical Society 2021, 143
(41)
, 17047-17058. https://doi.org/10.1021/jacs.1c06559
- Michaela Pieszka, Shen Han, Christiane Volkmann, Robert Graf, Ingo Lieberwirth, Katharina Landfester, David Y. W. Ng, Tanja Weil. Controlled Supramolecular Assembly Inside Living Cells by Sequential Multistaged Chemical Reactions. Journal of the American Chemical Society 2020, 142
(37)
, 15780-15789. https://doi.org/10.1021/jacs.0c05261
- Min Su, Tao Peng, Yingdi Zhu, Juan Li. Nucleic Acid Covalent Tags. ChemBioChem 2025, 26
(5)
https://doi.org/10.1002/cbic.202400805
- Yong Ke, Shuangxiu Li, Yan Shao, Qiushi Li, Ying Wang, Zhanzhan Zhang, Yang Liu. Calixarene Modification Strategy for Efficient Intracellular Protein Delivery. Macromolecular Bioscience 2025, 11 https://doi.org/10.1002/mabi.202400626
- Mengmeng Zheng, Lingchao Kong, Jianmin Gao. Boron enabled bioconjugation chemistries. Chemical Society Reviews 2024, 53
(24)
, 11888-11907. https://doi.org/10.1039/D4CS00750F
- Kevin Herr, Mark V. Höfler, Henrike Heise, Fabien Aussenac, Felix Kornemann, David Rosenberger, Martin Brodrecht, Marcos de Oliveira, Gerd Buntkowsky, Torsten Gutmann. Biradicals based on PROXYL containing building blocks for efficient dynamic nuclear polarization in biotolerant media. Journal of Magnetic Resonance Open 2024, 20 , 100152. https://doi.org/10.1016/j.jmro.2024.100152
- Ferran Esteve, Tanguy Rieu, Jean-Marie Lehn. Key structural features to favour imines over hydrates in water: pyridoxal phosphate as a muse. Chemical Science 2024, 15
(27)
, 10408-10415. https://doi.org/10.1039/D4SC02206H
- Dominik Schauenburg, Tanja Weil. Chemical Reactions in Living Systems. Advanced Science 2024, 11
(8)
https://doi.org/10.1002/advs.202303396
- Preeti Chauhan, Ragendu V., Mohan Kumar, Rajib Molla, Surya Dev Mishra, Sneha Basa, Vishal Rai. Chemical technology principles for selective bioconjugation of proteins and antibodies. Chemical Society Reviews 2024, 53
(1)
, 380-449. https://doi.org/10.1039/D3CS00715D
- Florian M. Zielke, Floris P. J. T. Rutjes. Recent Advances in Bioorthogonal Ligation and Bioconjugation. Topics in Current Chemistry 2023, 381
(6)
https://doi.org/10.1007/s41061-023-00445-6
- Katerina Gavriel, Dustin C. A. van Doeselaar, Daniëlle W. T. Geers, Kevin Neumann. Click’n lock
: rapid exchange between unsymmetric tetrazines and thiols for reversible, chemoselective functionalisation of biomolecules with on-demand bioorthogonal locking. RSC Chemical Biology 2023, 4
(9)
, 685-691. https://doi.org/10.1039/D3CB00062A
- Selina Hollstein, Philipp Erdmann, Andreas Ulmer, Henrik Löw, Lutz Greb, Max von Delius. Trialkoxysilane Exchange: Scope, Mechanism, Cryptates and pH‐Response. Angewandte Chemie 2023, 135
(26)
https://doi.org/10.1002/ange.202304083
- Selina Hollstein, Philipp Erdmann, Andreas Ulmer, Henrik Löw, Lutz Greb, Max von Delius. Trialkoxysilane Exchange: Scope, Mechanism, Cryptates and pH‐Response. Angewandte Chemie International Edition 2023, 62
(26)
https://doi.org/10.1002/anie.202304083
- Samuel L. Scinto, Tyler R. Reagle, Joseph M. Fox. Affinity Bioorthogonal Chemistry (ABC) Tags for Site‐Selective Conjugation, On‐Resin Protein‐Protein Coupling, and Purification of Protein Conjugates. Angewandte Chemie 2022, 134
(45)
https://doi.org/10.1002/ange.202207661
- Samuel L. Scinto, Tyler R. Reagle, Joseph M. Fox. Affinity Bioorthogonal Chemistry (ABC) Tags for Site‐Selective Conjugation, On‐Resin Protein‐Protein Coupling, and Purification of Protein Conjugates. Angewandte Chemie International Edition 2022, 61
(45)
https://doi.org/10.1002/anie.202207661
- Marco Raabe, Astrid Johanna Heck, Siska Führer, Dominik Schauenburg, Michaela Pieszka, Tao Wang, Maksymilian Marek Zegota, Lutz Nuhn, David Y. W. Ng, Seah Ling Kuan, Tanja Weil. Assembly of pH‐Responsive Antibody‐Drug‐Inspired Conjugates. Macromolecular Bioscience 2022, 22
(2)
https://doi.org/10.1002/mabi.202100299
- Fatima Abi-Ghaida. The serendipitous integration of small boron-embedded molecules into medicinal chemistry. 2022, 321-410. https://doi.org/10.1016/B978-0-12-822127-3.00006-5
- Kevin Herr, Max Fleckenstein, Martin Brodrecht, Mark V. Höfler, Henrike Heise, Fabien Aussenac, Torsten Gutmann, Michael Reggelin, Gerd Buntkowsky. A novel strategy for site selective spin-labeling to investigate bioactive entities by DNP and EPR spectroscopy. Scientific Reports 2021, 11
(1)
https://doi.org/10.1038/s41598-021-92975-6
- Lujuan Xu, Maria J. S. A. Silva, Pedro M. P. Gois, Seah Ling Kuan, Tanja Weil. Chemoselective cysteine or disulfide modification
via
single atom substitution in chloromethyl acryl reagents. Chemical Science 2021, 12
(40)
, 13321-13330. https://doi.org/10.1039/D1SC03250J
- Shu Song, Ping Gao, Lin Sun, Dongwei Kang, Jacob Kongsted, Vasanthanathan Poongavanam, Peng Zhan, Xinyong Liu. Recent developments in the medicinal chemistry of single boron atom-containing compounds. Acta Pharmaceutica Sinica B 2021, 11
(10)
, 3035-3059. https://doi.org/10.1016/j.apsb.2021.01.010
- Corey. A. Stevens, Kuljeet Kaur, Harm-Anton Klok. Self-assembly of protein-polymer conjugates for drug delivery. Advanced Drug Delivery Reviews 2021, 174 , 447-460. https://doi.org/10.1016/j.addr.2021.05.002
- Reinder H. de Vries, Jakob H. Viel, Oscar P. Kuipers, Gerard Roelfes. Rapid and Selective Chemical Editing of Ribosomally Synthesized and Post‐Translationally Modified Peptides (RiPPs) via Cu
II
‐Catalyzed β‐Borylation of Dehydroamino Acids. Angewandte Chemie 2021, 133
(8)
, 3992-3996. https://doi.org/10.1002/ange.202011460
- Reinder H. de Vries, Jakob H. Viel, Oscar P. Kuipers, Gerard Roelfes. Rapid and Selective Chemical Editing of Ribosomally Synthesized and Post‐Translationally Modified Peptides (RiPPs) via Cu
II
‐Catalyzed β‐Borylation of Dehydroamino Acids. Angewandte Chemie International Edition 2021, 60
(8)
, 3946-3950. https://doi.org/10.1002/anie.202011460
- Saurav Chatterjee, Eric V. Anslyn, Anupam Bandyopadhyay. Boronic acid based dynamic click chemistry: recent advances and emergent applications. Chemical Science 2021, 12
(5)
, 1585-1599. https://doi.org/10.1039/D0SC05009A
- Seah Ling Kuan, Marco Raabe. Solid‐Phase Protein Modifications: Towards Precision Protein Hybrids for Biological Applications. ChemMedChem 2021, 16
(1)
, 94-104. https://doi.org/10.1002/cmdc.202000412
- Guangzhong Yin, Jingjing Wei, Yu Shao, Wen-Hao Wu, Lianjie Xu, Wen-Bin Zhang. Native conjugation between proteins and [60]fullerene derivatives using SpyTag as a reactive handle. Chinese Chemical Letters 2021, 32
(1)
, 353-356. https://doi.org/10.1016/j.cclet.2020.04.034
- Landa Purushottam, Unnikrishnan V. B., Dattatraya Gautam Rawale, Mansi Gujrati, Surya Dev Mishra, Sajeev T. K., Neelesh C. Reddy, Srinivasa Rao Adusumalli, Ram Kumar Mishra, Vishal Rai. A single amino acid Gly-tag enables metal-free protein purification. Chemical Science 2020, 11
(48)
, 13137-13142. https://doi.org/10.1039/D0SC01153C
- Roberto Russo, Rita Padanha, Fábio Fernandes, Luis F. Veiros, Francisco Corzana, Pedro M. P. Gois. Engineering Boron Hot Spots for the Site‐Selective Installation of Iminoboronates on Peptide Chains. Chemistry – A European Journal 2020, 26
(66)
, 15226-15231. https://doi.org/10.1002/chem.202002675
- Adam A. Caparco, Bettina R. Bommarius, Andreas S. Bommarius, Julie A. Champion. Protein‐inorganic calcium‐phosphate supraparticles as a robust platform for enzyme co‐immobilization. Biotechnology and Bioengineering 2020, 117
(7)
, 1979-1989. https://doi.org/10.1002/bit.27348
- Jiaming Zhuang, Bo Zhao, Xiangxi Meng, Jessica D. Schiffman, Sarah L. Perry, Richard W. Vachet, S. Thayumanavan. A programmable chemical switch based on triggerable Michael acceptors. Chemical Science 2020, 11
(8)
, 2103-2111. https://doi.org/10.1039/C9SC05841A
- Christina Picken, Sahar Awwad, Mire Zloh, Hanieh Khalili, Steve Brocchini. Protein modification by bis-alkylation. 2020, 351-385. https://doi.org/10.1016/B978-0-444-64081-9.00016-4
- Cecilia Samaniego Lopez, Jimena H. Martínez, Sofía L. Acebedo, Carla C. Spagnuolo. Benzoxaboroles as dynamic covalent receptors for bioconjugation and transport of nucleosides and related drugs: Proof of action in HeLa cells. Bioorganic Chemistry 2019, 90 , 103059. https://doi.org/10.1016/j.bioorg.2019.103059
- João P. M. António, Roberto Russo, Cátia Parente Carvalho, Pedro M. S. D. Cal, Pedro M. P. Gois. Boronic acids as building blocks for the construction of therapeutically useful bioconjugates. Chemical Society Reviews 2019, 48
(13)
, 3513-3536. https://doi.org/10.1039/C9CS00184K
- Marco Lucchino, Anne Billet, Antoine Versini, Harikrishna Bavireddi, Bhanu‐Das Dasari, Sylvain Debieu, Ludovic Colombeau, Tatiana Cañeque, Alain Wagner, Géraldine Masson, Frédéric Taran, Philippe Karoyan, Muriel Delepierre, Christine Gaillet, Anne Houdusse, Sébastien Britton, Frédéric Schmidt, Jean‐Claude Florent, Philippe Belmont, David Monchaud, Janine Cossy, Christophe Thomas, Arnaud Gautier, Ludger Johannes, Raphaël Rodriguez. 2nd PSL Chemical Biology Symposium (2019): At the Crossroads of Chemistry and Biology. ChemBioChem 2019, 20
(7)
, 968-973. https://doi.org/10.1002/cbic.201900092
- Emily A. Hoyt, Pedro M. S. D. Cal, Bruno L. Oliveira, Gonçalo J. L. Bernardes. Contemporary approaches to site-selective protein modification. Nature Reviews Chemistry 2019, 3
(3)
, 147-171. https://doi.org/10.1038/s41570-019-0079-1
- Seah Ling Kuan, Fernando R. G. Bergamini, Tanja Weil. Functional protein nanostructures: a chemical toolbox. Chemical Society Reviews 2018, 47
(24)
, 9069-9105. https://doi.org/10.1039/C8CS00590G
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Scheme 1
Scheme 1. Synthesis of Boronic Acid Intercalator (BA-IC)aaa. methacryloyl chloride, Et3N, CH2Cl2, 99%; b. 1. I2, sodium tosylate, CH2Cl2; 2. Et3N, EA, 52%; c. TFA, CH2Cl2, 99%; d. HBTU, DIEA, 3-aminophenylboronic acid hydrochloride, DMF, 65%.
Scheme 2
Scheme 2. Incorporation of BA-IC into the LysozymeFigure 1
Figure 1. FPLC chromatogram of the reaction mixture with visible separation of lysozyme, modified lysozyme, and a higher molecular weight side product (retention volume = 19.82 mL).
Figure 2
Figure 2. (a) MALDI-TOF-MS interpretation of BA-lysozyme digest. Peak corresponding to modified fragment was selected for MSMS. 2,5-Dihydrobenzoic acid (DHB) was used as a matrix. (b) Interpretation of BA modified fragment’s MSMS spectrum. To simplify, only a, b, and y ion series for 6–13 fragment are annotated. Annotations for a-17, b-17 ion series and 126–128 fragment are included in the Supporting Information.
Figure 3
Figure 3. Rate of decrease in absorbance (slope) is proportional to lysozyme activity. Clearance of cell suspension turbidity proceeds with similar pace in case of both BA-modified (orange) and native lysozyme (red). Data are plotted as n = 4 ± standard errors of the mean (SEM).
Scheme 3
Scheme 3. Synthesis of SHA-BDPaaa. CuSO4, sodium ascorbate, DMSO, 93%; b. BF3OEt2, MeOH/CH2Cl2, 76%.
Scheme 4
Scheme 4. Dynamic Covalent Bioconjugation of BA-Lysozyme with SHA-BDP and pH-Induced Cleavage of the BioconjugateFigure 4
Figure 4. Microscale thermophoresis confirms assembly of BA-lysozyme with SHA-BDP (blue line) and release of SHA-BDP upon acidification (gray dots). No binding was detected for the control (red and green dots). Data are plotted as n = 3 ± SEM.
References
This article references 53 other publications.
- 1Dozier, J. K. and Distefano, M. D. (2015) Site-Specific PEGylation of Therapeutic Proteins. Int. J. Mol. Sci. 16, 25831– 25864, DOI: 10.3390/ijms1610258311Site-specific pegylation of therapeutic proteinsDozier, Jonathan K.; Distefano, Mark D.International Journal of Molecular Sciences (2015), 16 (10), 25831-25864CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)The use of proteins as therapeutics has a long history and is becoming ever more common in modern medicine. While the no. of protein-based drugs is growing every year, significant problems still remain with their use. Among these problems are rapid degrdn. and excretion from patients, thus requiring frequent dosing, which in turn increases the chances for an immunol. response as well as increasing the cost of therapy. One of the main strategies to alleviate these problems is to link a polyethylene glycol (PEG) group to the protein of interest. This process, called PEGylation, has grown dramatically in recent years resulting in several approved drugs. Installing a single PEG chain at a defined site in a protein is challenging. Recently, there is has been considerable research into various methods for the site-specific PEGylation of proteins. This review seeks to summarize that work and provide background and context for how site-specific PEGylation is performed. After introducing the topic of site-specific PEGylation, recent developments using chem. methods are described. That is followed by a more extensive discussion of bioorthogonal reactions and enzymic labeling.
- 2Krall, N., da Cruz, F. P., Boutureira, O., and Bernardes, G. J. (2016) Site-selective protein-modification chemistry for basic biology and drug development. Nat. Chem. 8, 103– 113, DOI: 10.1038/nchem.23932Site-selective protein-modification chemistry for basic biology and drug developmentKrall, Nikolaus; da Cruz, Filipa P.; Boutureira, Omar; Bernardes, Goncalo J. L.Nature Chemistry (2016), 8 (2), 103-113CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. Nature has produced intricate machinery to covalently diversify the structure of proteins after their synthesis in the ribosome. In an attempt to mimic nature, chemists have developed a large set of reactions that enable post-expression modification of proteins at pre-detd. sites. These reactions are now used to selectively install particular modifications on proteins for many biol. and therapeutic applications. For example, they provide an opportunity to install post-translational modifications on proteins to det. their exact biol. roles. Labeling of proteins in live cells with fluorescent dyes allows protein uptake and intracellular trafficking to be tracked and also enables physiol. parameters to be measured optically. Through the conjugation of potent cytotoxicants to antibodies, novel anti-cancer drugs with improved efficacy and reduced side effects may be obtained. In this Perspective, we highlight the most exciting current and future applications of chem. site-selective protein modification and consider which hurdles still need to be overcome for more widespread use.
- 3Kuan, S. L., Wang, T., and Weil, T. (2016) Site-Selective Disulfide Modification of Proteins: Expanding Diversity beyond the Proteome. Chem. - Eur. J. 22, 17112– 17129, DOI: 10.1002/chem.2016022983Site-Selective Disulfide Modification of Proteins: Expanding Diversity beyond the ProteomeKuan, Seah Ling; Wang, Tao; Weil, TanjaChemistry - A European Journal (2016), 22 (48), 17112-17129CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The synthetic transformation of polypeptides with mol. accuracy holds great promise for providing functional and structural diversity beyond the proteome. Consequently, the last decade has seen an exponential growth of site-directed chem. to install addnl. features into peptides and proteins even inside living cells. The disulfide rebridging strategy has emerged as a powerful tool for site-selective modifications since most proteins contain disulfide bonds. In this Review, we present the chem. design, advantages and limitations of the disulfide rebridging reagents, while summarizing their relevance for synthetic customization of functional protein bioconjugates, as well as the resultant impact and advancement for biomedical applications.
- 4Cho, H., Daniel, T., Buechler, Y. J., Litzinger, D. C., Maio, Z., Putnam, A.-M. H., Kraynov, V. S., Sim, B.-C., Bussell, S., Javahishvili, T., Kaphle, S., Viramontes, G., Ong, M., Chu, S., Becky, G. C., Lieu, R., Knudsen, N., Castiglioni, P., Norman, T. C., Axelrod, D. W., Hoffman, A. R., Schultz, P. G., DiMarchi, R. D., and Kimmel, B. E. (2011) Optimized clinical performance of growth hormone with an expanded genetic code. Proc. Natl. Acad. Sci. U. S. A. 108, 9060– 5, DOI: 10.1073/pnas.11003871084Optimized clinical performance of growth hormone with an expanded genetic codeCho, Ho; Daniel, Tom; Buechler, Ying Ji; Litzinger, David C.; Maio, Zhenwei; Putnam, Anna-Maria Hays; Kraynov, Vadim S.; Sim, Bee-Cheng; Bussell, Stuart; Javahishvili, Tsotne; Kaphle, Sami; Viramontes, Guillermo; Ong, Mike; Chu, Stephanie; Becky, G. C.; Lieu, Ricky; Knudsen, Nick; Castiglioni, Paola; Norman, Thea C.; Axelrod, Douglas W.; Hoffman, Andrew R.; Schultz, Peter G.; DiMarchi, Richard D.; Kimmel, Bruce E.Proceedings of the National Academy of Sciences of the United States of America (2011), 108 (22), 9060-9065, S9060/1-S9060/3CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The ribosomal incorporation of nonnative amino acids into polypeptides in living cells provides the opportunity to endow therapeutic proteins with unique pharmacol. properties. We report here the first clin. study of a biosynthetic protein produced using an expanded genetic code. Incorporation of p-acetylphenylalanine (pAcF) at distinct locations in human growth hormone (hGH) allowed site-specific conjugation with polyethylene glycol (PEG) to produce homogeneous hGH variants. A mono-PEGylated mutant hGH modified at residue 35 demonstrated favorable pharmacodynamic properties in GH-deficient rats. Clin. studies in GH-deficient adults demonstrated efficacy and safety comparable to native human growth hormone therapy but with increased potency and reduced injection frequency. This example illustrates the utility of nonnative amino acids to optimize protein therapeutics in an analogous fashion to the use of medicinal chem. to optimize conventional natural products, low mol. wt. drugs, and peptides.
- 5Elliott, T. S., Townsley, F. M., Bianco, A., Ernst, R. J., Sachdeva, A., Elsässer, S. J., Davis, L., Lang, K., Pisa, R., Greiss, S., Lilley, K. S., and Chin, J. W. (2014) Proteome labeling and protein identification in specific tissues and at specific developmental stages in an animal. Nat. Biotechnol. 32, 465– 472, DOI: 10.1038/nbt.28605Proteome labeling and protein identification in specific tissues and at specific developmental stages in an animalElliott, Thomas S.; Townsley, Fiona M.; Bianco, Ambra; Ernst, Russell J.; Sachdeva, Amit; Elsaesser, Simon J.; Davis, Lloyd; Lang, Kathrin; Pisa, Rudolf; Greiss, Sebastian; Lilley, Kathryn S.; Chin, Jason W.Nature Biotechnology (2014), 32 (5), 465-472CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Identifying the proteins synthesized at specific times in cells of interest in an animal will facilitate the study of cellular functions and dynamic processes. Here we introduce stochastic orthogonal recoding of translation with chemoselective modification (SORT-M) to address this challenge. SORT-M involves modifying cells to express an orthogonal aminoacyl-tRNA synthetase/tRNA pair to enable the incorporation of chem. modifiable analogs of amino acids at diverse sense codons in cells in rich media. We apply SORT-M to Drosophila melanogaster fed std. food to label and image proteins in specific tissues at precise developmental stages with diverse chemistries, including cyclopropene-tetrazine inverse electron demand Diels-Alder cycloaddn. reactions. We also use SORT-M to identify proteins synthesized in germ cells of the fly ovary without dissection. SORT-M will facilitate the definition of proteins synthesized in specific sets of cells to study development, and learning and memory in flies, and may be extended to other animals.
- 6Boutureira, O. and Bernardes, G. J. L. (2015) Advances in chemical protein modification. Chem. Rev. 115, 2174– 2195, DOI: 10.1021/cr500399p6Advances in Chemical Protein ModificationBoutureira, Omar; Bernardes, Goncalo J. L.Chemical Reviews (Washington, DC, United States) (2015), 115 (5), 2174-2195CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Transition metal-free and -mediated approaches are covered.
- 7Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., Chen, Y., Simpson, M., Tsai, S. P., Dennis, M. S., Lu, Y., Meng, Y. G., Ng, C., Yang, J., Lee, C. C., Duenas, E., Gorrell, J., Katta, V., Kim, A., McDorman, K., Flagella, K., Venook, R., Ross, S., Spencer, S. D., Lee Wong, W., Lowman, H. B., Vandlen, R., Sliwkowski, M. X., Scheller, R. H., Polakis, P., and Mallet, W. (2008) Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat. Biotechnol. 26, 925– 932, DOI: 10.1038/nbt.14807Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic indexJunutula, Jagath R.; Raab, Helga; Clark, Suzanna; Bhakta, Sunil; Leipold, Douglas D.; Weir, Sylvia; Chen, Yvonne; Simpson, Michelle; Tsai, Siao Ping; Dennis, Mark S.; Lu, Yanmei; Meng, Y. Gloria; Ng, Carl; Yang, Jihong; Lee, Chien C.; Duenas, Eileen; Gorrell, Jeffrey; Katta, Viswanatham; Kim, Amy; McDorman, Kevin; Flagella, Kelly; Venook, Rayna; Ross, Sarajane; Spencer, Susan D.; Wong, Wai Lee; Lowman, Henry B.; Vandlen, Richard; Sliwkowski, Mark X.; Scheller, Richard H.; Polakis, Paul; Mallet, WilliamNature Biotechnology (2008), 26 (8), 925-932CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogeneous conjugates with relatively narrow therapeutic index (max. tolerated dose/curative dose). Using leads from the authors' previously described phage display-based method to predict suitable conjugation sites, the authors engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb Ig folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepd. by conventional approaches. The favorable in vivo properties of the near-homogeneous compn. of this conjugate suggest that this strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.
- 8Stephan, J.-P., Chan, P., Lee, C., Nelson, C., Elliott, J. M., Bechtel, C., Raab, H., Xie, D., Akutagawa, J., Baudys, J., Saad, O., Prabhu, S., Wong, W. L. T., Vandlen, R., Jacobson, F., and Ebens, A. (2008) Anti-CD22-MCC-DM1 and MC-MMAF Conjugates: Impact of Assay Format on Pharmacokinetic Parameters Determination. Bioconjugate Chem. 19, 1673– 1683, DOI: 10.1021/bc800059t8Anti-CD22-MCC-DM1 and MC-MMAF Conjugates: Impact of Assay Format on Pharmacokinetic Parameters DeterminationStephan, Jean-Philippe; Chan, Pamela; Lee, Chien; Nelson, Christopher; Elliott, James Michael; Bechtel, Charity; Raab, Helga; Xie, David; Akutagawa, Jon; Baudys, Jakub; Saad, Ola; Prabhu, Saileta; Wong, Wai Lee T.; Vandlen, Richard; Jacobson, Fred; Ebens, AllenBioconjugate Chemistry (2008), 19 (8), 1673-1683CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)CD22 represents a promising target for antibody-drug conjugate therapy in the context of B cell malignancies since it rapidly internalizes, importing specifically bound antibodies with it. To det. the pharmacokinetic parameters of anti-CD22-MCC-DM1 and MC-MMAF conjugates, various approaches to quantifying total and conjugated antibody were investigated. Although the total antibody assay formats gave similar results for both conjugates, the mouse pharmacokinetic profile for the anti-CD22-MCC-DM1 and MC-MMAF appeared significantly different depending on the conjugated antibody assay format. Since these differences significantly impacted the PK parameters detn., we investigated the effect of the drug/antibody ratio on the total and conjugated antibody quantification using multiple assay formats. Our investigations revealed the limitations of some assay formats to quantify anti-CD22-MCC-DM1 and MC-MMAF with different drug load and in the context of a heterogeneous ADC population highlight the need to carefully plan the assay strategy for the total and conjugated antibody quantification in order to accurately det. the ADC PK parameters.
- 9Chalker, J. M., Bernardes, G. J. L., and Davis, B. G. (2011) A “Tag-and-Modify” Approach to Site-Selective Protein Modification. Acc. Chem. Res. 44, 730– 741, DOI: 10.1021/ar200056q9A "Tag-and-Modify" Approach to Site-Selective Protein ModificationChalker, Justin M.; Bernardes, Goncalo J. L.; Davis, Benjamin G.Accounts of Chemical Research (2011), 44 (9), 730-741CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Covalent modification can expand a protein's functional capacity. Fluorescent or radioactive labeling, for instance, allows imaging of a protein in real time. Labeling with an affinity probe enables isolation of target proteins and other interacting mols. At the other end of this functional spectrum, protein structures can be naturally altered by enzymic action. Protein-protein interactions, genetic regulation, and a range of cellular processes are under the purview of these post-translational modifications. The ability of protein chemists to install these covalent addns. selectively has been crit. for elucidating their roles in biol. Frequently the transformations must be applied in a site-specific manner, which demands the most selective chem. In this Account, we discuss the development and application of such chem. in our lab. A centerpiece of our strategy is a "tag-and-modify" approach, which entails sequential installation of a uniquely reactive chem. group into the protein (the "tag") and the selective or specific modification of this group. The chem. tag can be a natural or unnatural amino acid residue. Of the natural residues, cysteine is the most widely used as a tag. Early work in our program focused on selective disulfide formation in the synthesis of glycoproteins. For certain applications, the susceptibility of disulfides to redn. was a limitation and prompted the development of several methods for the synthesis of more stable thioether modifications. The desulfurization of disulfides and conjugate addn. to dehydroalanine are two routes to these modifications. The dehydroalanine tag has since proven useful as a general precursor to many modifications after conjugate addn. of various nucleophiles; phosphorylated, glycosylated, peptidylated, prenylated, and even mimics of methylated and acetylated lysine-contg. proteins are all accessible from dehydroalanine. While cysteine is a useful tag for selective modification, unnatural residues present the opportunity for bio-orthogonal chem. Azide-, arylhalide-, alkyne-, and alkene-contg. amino acids can be incorporated into proteins genetically and can be specifically modified through various transformations. These transformations often rely on metal catalysis. The Cu-catalyzed azide-alkyne addn., Ru-catalyzed olefin metathesis, and Pd-catalyzed cross-coupling are examples of such transformations. In the course of adapting these reactions to protein modification, we learned much about the behavior of these reactions in water, and in some cases entirely new catalysts were developed. Through a combination of these bio-orthogonal transformations from the panel of tag-and-modify reactions, multiple and distinct modifications can be installed on protein surfaces. Multiple modifications are common in natural systems, and synthetic access to these proteins has enabled study of their biol. role. Throughout these investigations, much has been learned in chem. and biol. The demands of selective protein modification have revealed many aspects of reaction mechanisms, which in turn have guided the design of reagents and catalysts that allow their successful deployment in water and in biol. milieu. With this ability to modify proteins, it is now possible to interrogate biol. systems with precision that was not previously possible.
- 10Zhang, X., Lu, W., Kwan, K., Bhattacharyya, D., and Wei, Y. (2017) Dual-Functional-Tag-Facilitated Protein Labeling and Immobilization. ACS Omega 2, 522– 528, DOI: 10.1021/acsomega.6b00512There is no corresponding record for this reference.
- 11Rybak, J.-N., Scheurer, S. B., Neri, D., and Elia, G. (2004) Purification of biotinylated proteins on streptavidin resin: A protocol for quantitative elution. Proteomics 4, 2296– 2299, DOI: 10.1002/pmic.20030078011Purification of biotinylated proteins on streptavidin resin: A protocol for quantitative elutionRybak, Jascha-N.; Scheurer, Simone B.; Neri, Dario; Elia, GiulianoProteomics (2004), 4 (8), 2296-2299CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)The interaction between streptavidin and biotin is one of the most widely used tools in chem. and biol. However, the release of biotinylated proteins from streptavidin resins remains a major problem, due to the extraordinary stability of this complex. We present a new protocol for the quant. elution of biotinylated proteins from streptavidin Sepharose, featuring harsh elution conditions and competition with free biotin. The usefulness of the method was demonstrated by the quant. recovery of biotinylated proteins from organ homogenates, obtained from mice perfused with a reactive ester deriv. of biotin.
- 12Campos, S. K., Parrott, M. B., and Barry, M. A. (2004) Avidin-based targeting and purification of a protein IX-modified, metabolically biotinylated adenoviral vector. Mol. Ther. 9, 942– 54, DOI: 10.1016/j.ymthe.2004.03.00612Avidin-based targeting and purification of a protein IX-modified, metabolically biotinylated adenoviral vectorCampos, Samuel K.; Parrott, M. Brandon; Barry, Michael A.Molecular Therapy (2004), 9 (6), 942-954CODEN: MTOHCK; ISSN:1525-0016. (Elsevier)While genetic modification of adenoviral vectors can produce vectors with modified tropism, incorporation of targeting peptides/proteins into the structural context of the virion can also result in destruction of ligand targeting or virion integrity. To combat this problem, the authors have developed a versatile targeting system using metabolically biotinylated adenoviral vectors bearing biotinylated fiber proteins. These vectors have been demonstrated to be useful as a platform for avidin-based ligand screening and vector targeting by conjugating biotinylated ligands to the virus using high-affinity tetrameric avidin (Kd = 10-15 M). The biotinylated vector could also be purified by biotin-reversible binding on monomeric avidin (Kd = 10-7 M). In this report, a second metabolically biotinylated adenovirus vector, Ad-IX-BAP, has been engineered by fusing a biotin acceptor peptide (BAP) to the C-terminus of the adenovirus pIX protein. This biotinylated vector displays twice as many biotins and was markedly superior for single-step affinity purifn. on monomeric avidin resin. However, unlike the fiber-biotinylated vector, Ad-IX-BAP failed to retarget to cells with biotinylated antibodies including anti-CD71 against the transferrin receptor. In contrast, Ad-IX-BAP was retargeted if transferrin, the cognate ligand for CD71, was used as a ligand rather than the anti-CD71. This work demonstrates the utility of metabolic biotinylation as a mol. screening tool to assess the utility of different viral capsid proteins for ligand display and the biol. and compatibility of different ligands and receptors for vector targeting applications. These results also demonstrate the utility of the pIX-biotinylated vector as a platform for gentle single-step affinity purifn. of adenoviral vectors.
- 13Bornhorst, J. A. and Falke, J. J. (2000) Purification of proteins using polyhistidine affinity tags. Methods Enzymol. 326, 245– 254, DOI: 10.1016/S0076-6879(00)26058-813Purification of proteins using polyhistidine affinity tagsBornhorst, Joshua A.; Falke, Joseph J.Methods in Enzymology (2000), 326 (Applications of Chimeric Genes and Hybrid Proteins, Pt. A), 245-254CODEN: MENZAU; ISSN:0076-6879. (Academic Press)A review with 30 refs. The expression and subsequent purifn. of recombinant proteins are widely employed in biochem. studies. A powerful purifn. method involves the use of peptide affinity tags, which are fused to the protein of interest and used to expedite protein purifn. via affinity chromatog. A widely employed method utilizes immobilized metal-affinity chromatog. (IMAC) to purify recombinant proteins contg. a short affinity tag consisting of polyhistidine residues. IMAC is a versatile method that can be utilized to rapidly purify polyhistidine affinity-tagged proteins, resulting in 100-fold enrichments in a single purifn. step. Affinity-tagged protein purities can be achieved at up to 95% purity by IMAC in high yield. Protocols for the method are described in detail. (c) 2000 Academic Press.
- 14Kimple, M. E., Brill, A. L., and Pasker, R. L. (2013) Overview of Affinity Tags for Protein Purification. Curr. Protoc Protein Sci. 73, 608– 616, DOI: 10.1002/0471140864.ps0909s73There is no corresponding record for this reference.
- 15Fexby, S. and Bü Low, L. (2004) Hydrophobic peptide tags as tools in bioseparation. Trends Biotechnol. 22, 511– 516, DOI: 10.1016/j.tibtech.2004.08.005There is no corresponding record for this reference.
- 16Jin, Y., Yu, C., Denman, R. J., and Zhang, W. (2013) Recent advances in dynamic covalent chemistry. Chem. Soc. Rev. 42, 6634– 6654, DOI: 10.1039/c3cs60044k16Recent advances in dynamic covalent chemistryJin, Yinghua; Yu, Chao; Denman, Ryan J.; Zhang, WeiChemical Society Reviews (2013), 42 (16), 6634-6654CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)A review. Dynamic covalent chem. (DCvC) has been strongly integrated into diverse research fields, and has enabled easy access to a variety of combinatorial libraries, 2-D macrocycles, and 3-D mol. cages that target many important applications, such as drug discovery, biotechnol., mol. sepn., light harvesting, etc. DCvC relies on the reversible formation and breaking of rather strong covalent bonding within mols. Therefore it combines the error-correction capability of supramol. chem. and the robustness of covalent bonding. Compared to those supramol. interactions, dynamic covalent reactions usually have slower kinetics and require the assistance of catalysts to achieve rapid equil. Although the scope of dynamic covalent reactions is rapidly expanding, the reversible reactions suitable for DCvC are still very limited. The identification and development of new dynamic reactions and catalysts would be crit. for the further advancement of DCvC. This review covers the recent development of dynamic covalent reactions as well as their applications.
- 17Bartolami, E., Bessin, Y., Gervais, V., Dumy, P., and Ulrich, S. (2015) Dynamic Expression of DNA Complexation with Self-assembled Biomolecular Clusters. Angew. Chem., Int. Ed. 54, 10183– 10187, DOI: 10.1002/anie.20150404717Dynamic Expression of DNA Complexation with Self-assembled Biomolecular ClustersBartolami, Eline; Bessin, Yannick; Gervais, Virginie; Dumy, Pascal; Ulrich, SebastienAngewandte Chemie, International Edition (2015), 54 (35), 10183-10187CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We report herein the implementation of a dynamic covalent chem. approach to the generation of multivalent clusters for DNA recognition. We show that biomol. clusters can be expressed in situ by a programmed self-assembly process using chemoselective ligations. The cationic clusters are shown, by fluorescence displacement assay, gel electrophoresis and isothermal titrn. calorimetry, to effectively complex DNA through multivalent interactions. The reversibility of the ligation was exploited to demonstrate that template effects occur, whereby DNA imposes component selection in order to favor the most active DNA-binding clusters. Furthermore, we show that a chem. effector can be used to trigger DNA release through component exchange reactions.
- 18Reuther, J. F., Dees, J. L., Kolesnichenko, I. V., Hernandez, E. T., Ukraintsev, D. V., Guduru, R., Whiteley, M., and Anslyn, E. V. (2017) Dynamic covalent chemistry enables formation of antimicrobial peptide quaternary assemblies in a completely abiotic manner. Nat. Chem. 10, 45– 50, DOI: 10.1038/nchem.2847There is no corresponding record for this reference.
- 19Diehl, K. L., Kolesnichenko, I. V., Robotham, S. A., Bachman, J. L., Zhong, Y., Brodbelt, J. S., and Anslyn, E. V. (2016) Click and chemically triggered declick reactions through reversible amine and thiol coupling via a conjugate acceptor. Nat. Chem. 8, 968– 973, DOI: 10.1038/nchem.260119Click and chemically triggered declick reactions through reversible amine and thiol coupling via a conjugate acceptorDiehl, Katharine L.; Kolesnichenko, Igor V.; Robotham, Scott A.; Bachman, J. Logan; Zhong, Ye; Brodbelt, Jennifer S.; Anslyn, Eric V.Nature Chemistry (2016), 8 (10), 968-973CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The coupling and decoupling of mol. units is a fundamental undertaking of org. chem. Herein we report the use of a very simple conjugate acceptor, derived from Meldrum's acid, for the sequential 'clicking' together of an amine and a thiol in aq. conditions at neutral pH. Subsequently, this linkage can be 'declicked' by a chem. trigger to release the original amine and thiol undisturbed. The reactivity differs from that of other crosslinking agents because the selectivity for sequential functionalization derives from an altering of the electrophilicity of the conjugate acceptor on the addn. of the amine. We describe the use of the procedure to modify proteins, create multicomponent libraries and synthesize oligomers, all of which can be declicked to their starting components in a controlled fashion when desired. Owing to the mild reaction conditions and ease of use in a variety of applications, the method is predicted to have wide utility.
- 20Wang, T., Ng, D. Y. W., Wu, Y., Thomas, J., TamTran, T., and Weil, T. (2014) Bis-sulfide bioconjugates for glutathione triggered tumor responsive drug release. Chem. Commun. 50, 1116– 1118, DOI: 10.1039/C3CC47003B20Bis-sulfide bioconjugates for glutathione triggered tumor responsive drug releaseWang, Tao; Ng, David Y. W.; Wu, Yuzhou; Thomas, Jessica; TamTran, Thuy; Weil, TanjaChemical Communications (Cambridge, United Kingdom) (2014), 50 (9), 1116-1118CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)The reaction of bis-sulfone conjugation reagents with disulfide bonds allows the site-specific modification of various peptides and proteins. Herein, we present the intracellular disintegration of bis-sulfide contg. somatostatin bioconjugates under controlled, tumor-relevant glutathione levels. GSH responsive release is demonstrated, which offers high potential for designing tumor responsive therapeutics.
- 21Brustad, E., Bushey, M. L., Lee, J. W., Groff, D., Liu, W., and Schultz, P. G. (2008) A Genetically Encoded Boronate-Containing Amino Acid. Angew. Chem., Int. Ed. 47, 8220– 8223, DOI: 10.1002/anie.20080324021A genetically encoded boronate-containing amino acidBrustad, Eric; Bushey, Mark L.; Lee, Jae Wook; Groff, Dan; Liu, Wenshe; Schultz, Peter G.Angewandte Chemie, International Edition (2008), 47 (43), 8220-8223CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)A biol. boronate: An orthogonal tRNA/aminoacyl-tRNA synthetase pair has been evolved for the genetic incorporation of a boronic acid into proteins. This amino acid has been used to purify proteins in a one-step scarless purifn. procedure as well as for the site-specific labeling of proteins using various boronic acid chemistries.
- 22Arzt, M., Seidler, C., Ng, D. Y. W., and Weil, T. (2014) Reversible Click Reactions with Boronic Acids to Build Supramolecular Architectures in Water. Chem. - Asian J. 9, 1994, DOI: 10.1002/asia.20140206122Reversible Click Reactions with Boronic Acids to Build Supramolecular Architectures in WaterArzt, Matthias; Seidler, Christiane; Ng, David Y. W.; Weil, TanjaChemistry - An Asian Journal (2014), 9 (8), 1994-2003CODEN: CAAJBI; ISSN:1861-4728. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The interaction of boronic acids with various bifunctional reagents offers great potential for the prepn. of responsive supramol. architectures. Boronic acids react with 1,2-diols yielding cyclic boronate esters that are stable at pH>7.4 but can be hydrolyzed at pH<5.0. The phenylboronic acid (PBA)-salicylhydroxamic acid (SHA) system offers ultra-fast reaction kinetics and high binding affinities. This Focus review summarizes the current advances in exploiting the bioorthogonal interaction of boronic acids to build pH-responsive supramol. architectures in water.
- 23Jackson, T. R., Springall, J. S., Rogalle, D., Masumoto, N., Ching Li, H., D’Hooge, F., Perera, S. P., Jenkins, T. A., James, T. D., Fossey, J. S., and van den Elsen, J. M. H. (2008) Boronate affinity saccharide electrophoresis: A novel carbohydrate analysis tool. Electrophoresis 29, 4185– 4191, DOI: 10.1002/elps.200800178There is no corresponding record for this reference.
- 24Ng, D. Y. W., Arzt, M., Wu, Y., Kuan, S. L., Lamla, M., and Weil, T. (2014) Constructing Hybrid Protein Zymogens through Protective Dendritic Assembly. Angew. Chem., Int. Ed. 53, 324– 328, DOI: 10.1002/anie.20130853324Constructing Hybrid Protein Zymogens through Protective Dendritic AssemblyNg, David Y. W.; Arzt, Matthias; Wu, Yuzhou; Kuan, Seah Ling; Lamla, Markus; Weil, TanjaAngewandte Chemie, International Edition (2014), 53 (1), 324-328CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The modulation of protein uptake and activity in response to physiol. changes forms an integral part of smart protein therapeutics. We describe herein the self-assembly of a pH-responsive dendrimer shell onto the surface of active enzymes (trypsin, papain, DNase I) as a supramol. protecting group to form a hybrid dendrimer-enzyme complex. The attachment is based on the interaction between boronic acid and salicyl hydroxamate, thus allowing the macromol. assembly to respond to changes in pH between 5.0 and 7.4 in a highly reversible fashion. Catalytic activity is efficiently blocked in the presence of the dendrimer shell but is quant. restored upon shell degrdn. under acidic conditions. Unlike the native proteases, the hybrid constructs are shown to be efficiently taken up by A549 cells and colocalized in the acidic compartments. The programmed intracellular release of the proteases induced cytotoxicity, thereby uncovering a new avenue for precision biotherapeutics.
- 25Seidler, C., Ng, D. Y. W., Wu, Y., and Weil, T. (2016) pH responsive supramolecular core-shell protein hybrids. Supramol. Chem. 28, 742– 746, DOI: 10.1080/10610278.2016.114076425pH responsive supramolecular core-shell protein hybridsSeidler, Christiane; Ng, David Y. W.; Wu, Yuzhou; Weil, TanjaSupramolecular Chemistry (2016), 28 (9-10), 742-746CODEN: SCHEER; ISSN:1029-0478. (Taylor & Francis Ltd.)PEGylation of proteins remains an integral part of macromol. therapeutics due to its well-known benign effects and pharmacokinetic enhancement properties. We report herein that PEGylation can be taken to the next level of complexity and dynamic behavior by introducing highly stable but responsive supramol. handles. By attaching small boronic acid groups onto proteins and salicylhydroxamate moiety to end-functionalise PEG chains, we demonstrate a comprehensive study on the facile assembly/disassembly of a core-shell protein-polymer architecture using fluorescence and microscale thermophoresis on a macromol. level. In addn., we demonstrate that both the activity and cellular transfer of functional proteins remained conserved throughout the assembly process thus establishing a rapid and orthogonal strategy towards protein PEGylation.
- 26Swaminathan, R., Ravi, V. K., Kumar, S., Kumar, M. V. S., and Chandra, N. (2011) Lysozyme: A model protein for amyloid research. Adv. Protein Chem. Struct. Biol. 84, 63– 111, DOI: 10.1016/B978-0-12-386483-3.00003-326Lysozyme: A model protein for amyloid researchSwaminathan, Rajaram; Ravi, Vijay Kumar; Kumar, Satish; Kumar, Mattaparthi Venkata Satish; Chandra, NividhAdvances in Protein Chemistry and Structural Biology (2011), 84 (), 63-111CODEN: APCSG7; ISSN:1876-1623. (Elsevier Ltd.)A review. Ever since lysozyme was discovered by Fleming in 1922, this protein has emerged as a model for investigations on protein structure and function. Over the years, several high-resoln. structures have yielded a wealth of structural data on this protein. Extensive studies on folding of lysozyme have shown how different regions of this protein dynamically interact with one another. Data is also available from numerous biotechnol. studies wherein lysozyme has been employed as a model protein for recovering active recombinant protein from inclusion bodies using small mols. like L-arginine. A variety of conditions have been developed in vitro to induce fibrillation in hen lysozyme. They include (a) acidic pH at elevated temp., (b) concd. solns. of ethanol, (c) moderate concns. of guanidinium hydrochloride at moderate temp., and (d) alk. pH at room temp. This review aims to bring together similarities and differences in aggregation mechanisms, morphol. of aggregates, and related issues that arise using the different conditions mentioned above to improve our understanding. The alk. pH condition (pH 12.2), discovered and studied extensively in our lab, shall receive special attention. More than a decade ago, it was revealed that mutations in human lysozyme can cause accumulation of large quantities of amyloid in liver, kidney, and other regions of gastrointestinal tract. Understanding the mechanism of lysozyme aggregation will probably have therapeutic implications for the treatment of systemic nonneuropathic amyloidosis. Numerous studies have begun to focus attention on inhibition of lysozyme aggregation using antibody or small mols. The enzymic activity of lysozyme presents a convenient handle to quantify the native population of lysozyme in a sample where aggregation has been inhibited. The rich information available on lysozyme coupled with the multiple conditions that have been successful in inducing/inhibiting its aggregation in vitro makes lysozyme an ideal model protein to investigate amyloidogenesis.
- 27Rambaran, R. N. and Serpell, L. C. (2008) Amyloid fibrils: abnormal protein assembly. Prion 2, 112– 7, DOI: 10.4161/pri.2.3.748827Amyloid fibrils: abnormal protein assemblyRambaran Roma N; Serpell Louise CPrion (2008), 2 (3), 112-7 ISSN:.Amyloid refers to the abnormal fibrous, extracellular, proteinaceous deposits found in organs and tissues. Amyloid is insoluble and is structurally dominated by beta-sheet structure. Unlike other fibrous proteins it does not commonly have a structural, supportive or motility role but is associated with the pathology seen in a range of diseases known as the amyloidoses. These diseases include Alzheimer's, the spongiform encephalopathies and type II diabetes, all of which are progressive disorders with associated high morbidity and mortality. Not surprisingly, research into the physicochemical properties of amyloid and its formation is currently intensely pursued. In this chapter we will highlight the key scientific findings and discuss how the stability of amyloid fibrils impacts on bionanotechnology.
- 28Mankar, S., Anoop, A., Sen, S., and Maji, S. K. (2011) Nanomaterials: amyloids reflect their brighter side. Nano Rev. 2, 6032, DOI: 10.3402/nano.v2i0.6032There is no corresponding record for this reference.
- 29Meier, C. and Welland, M. E. (2011) Wet-Spinning of Amyloid Protein Nanofibers into Multifunctional High-Performance Biofibers. Biomacromolecules 12, 3453– 3459, DOI: 10.1021/bm200575229Wet-Spinning of Amyloid Protein Nanofibers into Multifunctional High-Performance BiofibersMeier, Christoph; Welland, Mark E.Biomacromolecules (2011), 12 (10), 3453-3459CODEN: BOMAF6; ISSN:1525-7797. (American Chemical Society)Amyloid nanofibers derived from hen egg white lysozyme were processed into macroscopic fibers in a wet-spinning process based on interfacial polyion complexation using a polyanionic polysaccharide as crosslinker. As a result of their amyloid nanostructure, the hierarchically self-assembled protein fibers have a stiffness of up to 14 GPa and a tensile strength of up to 326 MPa. Fine-tuning of the polyelectrolytic interactions via pH allows to trigger the release of small mols., as demonstrated with riboflavin-5'-phosphate. The amyloid fibrils, highly oriented within the gellan gum matrix, were mineralized with calcium phosphate, mimicking the fibrolamellar structure of bone. The formed mineral crystals are highly oriented along the nanofibers, thus resulting in a 9-fold increase in fiber stiffness.
- 30Wang, T., Riegger, A., Lamla, M., Wiese, S., Oeckl, P., Otto, M., Wu, Y., Fischer, S., Barth, H., Kuan, S. L., and Weil, T. (2016) Water-soluble allyl sulfones for dual site-specific labelling of proteins and cyclic peptides. Chem. Sci. 7, 3234– 3239, DOI: 10.1039/C6SC00005C30Water-soluble allyl sulfones for dual site-specific labelling of proteins and cyclic peptidesWang, Tao; Riegger, Andreas; Lamla, Markus; Wiese, Sebastian; Oeckl, Patrick; Otto, Markus; Wu, Yuzhou; Fischer, Stephan; Barth, Holger; Kuan, Seah Ling; Weil, TanjaChemical Science (2016), 7 (5), 3234-3239CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Water-sol. allyl sulfones provide convenient site-specific disulfide rebridging of native proteins and cyclic peptides. The site-selective functionalization of (a) the peptide hormone somatostatin, (b) the interchain disulfide of bovine insulin and (c) functionalization of the proteins GFP and lysozyme with allyl sulfones proceeds in aq. soln. Allyl sulfones offer three functionalizable sites that react with thiol contg. mols. in a step-wise fashion. Dual labeling of proteins and cyclic peptides is achieved attachment of chromophore and affinity tag in a single reaction step, which is of great significance for the construction of precise multifunctional peptide and protein conjugates.
- 31Brocchini, S., Balan, S., Godwin, A., Choi, J.-W., Zloh, M., and Shaunak, S. (2006) PEGylation of native disulfide bonds in proteins. Nat. Chem. Biol. 1, 2241– 2252, DOI: 10.1038/nprot.2006.346There is no corresponding record for this reference.
- 32Robinson, E., Nunes, J. P. M., Vassileva, V., Maruani, A., Nogueira, J. C. F., Smith, M. E. B., Pedley, R. B., Caddick, S., Baker, J. R., and Chudasama, V. (2017) Pyridazinediones deliver potent, stable, targeted and efficacious antibody–drug conjugates (ADCs) with a controlled loading of 4 drugs per antibody. RSC Adv. 7, 9073– 9077, DOI: 10.1039/C7RA00788D32Pyridazinediones deliver potent, stable, targeted and efficacious antibody-drug conjugates (ADCs) with a controlled loading of 4 drugs per antibodyRobinson, Eifion; Nunes, Joao P. M.; Vassileva, Vessela; Maruani, Antoine; Nogueira, Joao C. F.; Smith, Mark E. B.; Pedley, R. Barbara; Caddick, Stephen; Baker, James R.; Chudasama, VijayRSC Advances (2017), 7 (15), 9073-9077CODEN: RSCACL; ISSN:2046-2069. (Royal Society of Chemistry)Herein we report the use of pyridazinediones to functionalize the native solvent accessible interstrand disulfide bonds in trastuzumab with monomethyl auristatin E (MMAE). This method of conjugation delivers serum stable antibody-drug conjugates (ADCs) with a controlled drug loading of 4. Moreover, we demonstrate that the MMAE-bearing ADCs are potent, selective and efficacious against cancer cell lines in both in vitro and in vivo models.
- 33Greene, M. K., Richards, D. A., Nogueira, J. C. F., Campbell, K., Smyth, P., Fernández, M., Scott, C. J., and Chudasama, V. (2018) Forming next-generation antibody–nanoparticle conjugates through the oriented installation of non-engineered antibody fragments. Chem. Sci. 9, 79– 87, DOI: 10.1039/C7SC02747H33Forming next-generation antibody-nanoparticle conjugates through the oriented installation of non-engineered antibody fragmentsGreene, Michelle K.; Richards, Daniel A.; Nogueira, Joao C. F.; Campbell, Katrina; Smyth, Peter; Fernandez, Marcos; Scott, Christopher J.; Chudasama, VijayChemical Science (2018), 9 (1), 79-87CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The successful development of targeted nanotherapeutics is contingent upon the conjugation of therapeutic nanoparticles to target-specific ligands, with particular emphasis being placed on antibody-based ligands. Thus, new methods that enable the covalent and precise installation of targeting antibodies to nanoparticle surfaces are greatly desired, esp. those which do not rely on costly and time-consuming antibody engineering techniques. Herein we present a novel method for the highly controlled and oriented covalent conjugation of non-engineered antibody F(ab) fragments to PLGA-PEG nanoparticles using disulfide-selective pyridazinedione linkers and strain-promoted alkyne-azide click chem. Exemplification of this method with trastuzumab and cetuximab showed significant improvements in both conjugation efficiency and antigen binding capability, when compared to commonly employed strategies for antibody-nanoparticle construction. This new approach paves the way for the development of antibody-targeted nanomedicines with improved paratope availability, reproducibility and uniformity to enhance both biol. activity and ease of manuf.
- 34Trivedi, M. V., Laurence, J. S., and Siahaan, T. J. (2009) The role of thiols and disulfides on protein stability. Curr. Protein Pept. Sci. 10, 614– 25, DOI: 10.2174/13892030978963053434The role of thiols and disulfides on protein stabilityTrivedi, Maulik V.; Laurence, Jennifer S.; Siahaan, Teruna J.Current Protein and Peptide Science (2009), 10 (6), 614-625CODEN: CPPSCM; ISSN:1389-2037. (Bentham Science Publishers Ltd.)A review. There has been a tremendous increase in the no. of approved drugs derived from recombinant proteins; however, their development as potential drugs has been hampered by their instability that causes difficulty to formulate them as therapeutic agents. It has been shown that the reactivity of thiol and disulfide functional groups could catalyze chem. (i.e., oxidn. and β-elimination reactions) and phys. (i.e., aggregation and pptn.) degrdn. of proteins. Because most proteins contain a free Cys residue and/or a disulfide bond, this review is focused on their roles in the phys. and chem. stability of proteins. The effect of introducing a disulfide bond to improve phys. stability of proteins and the mechanisms of degrdn. of disulfide bond were discussed. The qual./quant. methods to det. the presence of the thiol in the Cys residue and various methods to derivatize the thiol group for improving protein stability were also illustrated.
- 35Rosen, C. B. and Francis, M. B. (2017) Targeting the N terminus for site-selective protein modification. Nat. Chem. Biol. 13, 697– 705, DOI: 10.1038/nchembio.241635Targeting the N terminus for site-selective protein modificationRosen, Christian B.; Francis, Matthew B.Nature Chemical Biology (2017), 13 (7), 697-705CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. The formation of well-defined protein bioconjugates is crit. for many studies and technologies in chem. biol. Tried-and-true methods for accomplishing this typically involve the targeting of cysteine residues, but the rapid growth of contemporary bioconjugate applications has required an expanded repertoire of modification techniques. One very powerful set of strategies involves the modification of proteins at their N termini, as these positions are typically solvent exposed and provide chem. distinct sites for many protein targets. Several chem. techniques can be used to modify N-terminal amino acids directly or convert them into unique functional groups for further ligations. A growing no. of N-terminus-specific enzymic ligation strategies provided addnl. possibilities. This Perspective provides an overview of N-terminal modification techniques and the chem. rationale governing each. Examples of specific N-terminal protein conjugates are provided, along with their uses in a no. of diverse biol. applications.
- 36Tanaka, K., Fukase, K., and Katsumura, S. (2011) Exploring a Unique Reactivity of 6p-Azaelectrocyclization to Enzyme Inhibition, Natural Products Synthesis, and Molecular Imaging: An Approach to Chemical Biology by Synthetic Chemists. Synlett 2011, 2115– 2139, DOI: 10.1055/s-0030-1261192There is no corresponding record for this reference.
- 37Ban, H., Nagano, M., Gavrilyuk, J., Hakamata, W., Inokuma, T., and Barbas, C. F. (2013) Facile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click Reaction. Bioconjugate Chem. 24, 520– 532, DOI: 10.1021/bc300665t37Facile and Stabile Linkages through Tyrosine: Bioconjugation Strategies with the Tyrosine-Click ReactionBan, Hitoshi; Nagano, Masanobu; Gavrilyuk, Julia; Hakamata, Wataru; Inokuma, Tsubasa; Barbas, Carlos F., IIIBioconjugate Chemistry (2013), 24 (4), 520-532CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The scope, chemoselectivity, and utility of the click-like tyrosine labeling reaction with 4-phenyl-3H-1,2,4-triazoline-3,5(4H)-diones (PTADs) is reported. To study the utility and chemoselectivity of PTAD derivs. in peptide and protein chem., we synthesized PTAD derivs. possessing azide, alkyne, and ketone groups and studied their reactions with amino acid derivs. and peptides of increasing complexity. With proteins we studied the compatibility of the tyrosine click reaction with cysteine and lysine-targeted labeling approaches and demonstrate that chemoselective trifunctionalization of proteins is readily achieved. In particular cases, we noted that PTAD decompn. resulted in formation of a putative isocyanate byproduct that was promiscuous in labeling. This side reaction product, however, was readily scavenged by the addn. of a small amt. of 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) to the reaction medium. To study the potential of the tyrosine click reaction to introduce poly(ethylene glycol) chains onto proteins (PEGylation), we demonstrate that this novel reagent provides for the selective PEGylation of chymotrypsinogen, whereas traditional succinimide-based PEGylation targeting lysine residues provided a more diverse range of PEGylated products. Finally, we applied the tyrosine click reaction to create a novel antibody-drug conjugate. For this purpose, we synthesized a PTAD deriv. linked to the HIV entry inhibitor aplaviroc. Labeling of the antibody trastuzumab with this reagent provided a labeled antibody conjugate that demonstrated potent HIV-1 neutralization activity demonstrating the potential of this reaction in creating protein conjugates with small mols. The tyrosine click linkage demonstrated stability to extremes of pH, temp., and exposure to human blood plasma indicating that this linkage is significantly more robust than maleimide-type linkages that are commonly employed in bioconjugations. These studies support the broad utility of this reaction in the chemoselective modification of small mols., peptides, and proteins under mild aq. conditions over a broad pH range using a wide variety of biol. acceptable buffers such as phosphate buffered saline (PBS) and 2-amino-2-hydroxymethyl-propane-1,3-diol (Tris) buffers as well as others and mixed buffered compns.
- 38Santos, F. M. F., Rosa, J. N., Candeias, N. R., Carvalho, C. P., Matos, A. I., Ventura, A. E., Florindo, H. F., Silva, L. C., Pischel, U., and Gois, P. M. P. (2016) A Three-Component Assembly Promoted by Boronic Acids Delivers a Modular Fluorophore Platform (BASHY Dyes). Chem. - Eur. J. 22, 1631– 1637, DOI: 10.1002/chem.20150394338A Three-Component Assembly Promoted by Boronic Acids Delivers a Modular Fluorophore Platform (BASHY Dyes)Santos, Fabio M. F.; Rosa, Joao N.; Candeias, Nuno R.; Carvalho, Catia Parente; Matos, Ana I.; Ventura, Ana E.; Florindo, Helena F.; Silva, Liana C.; Pischel, Uwe; Gois, Pedro M. P.Chemistry - A European Journal (2016), 22 (5), 1631-1637CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)The modular assembly of boronic acids with Schiff-base ligands enabled the construction of innovative fluorescent dyes [boronic acid salicylidenehydrazone (BASHY)] with suitable structural and photophys. properties for live cell bioimaging applications. This reaction enabled the straightforward synthesis (yields up to 99 %) of structurally diverse and photostable dyes that exhibit a polarity-sensitive green-to-yellow emission with high quantum yields of up to 0.6 in nonpolar environments. These dyes displayed a high brightness (up to 54 000 M-1 cm-1). The promising structural and fluorescence properties of BASHY dyes fostered the prepn. of non-cytotoxic, stable, and highly fluorescent poly(lactide-co-glycolide) nanoparticles that were effectively internalized by dendritic cells. The dyes were also shown to selectively stain lipid droplets in HeLa cells, without inducing any appreciable cytotoxicity or competing plasma membrane labeling; this confirmed their potential as fluorescent stains.
- 39Cal, P. M. S. D., Sieglitz, F., Santos, F. M. F., Parente Carvalho, C., Guerreiro, A., Bertoldo, J. B., Pischel, U., Gois, P. M. P., and Bernardes, G. J. L. (2017) Site-selective installation of BASHY fluorescent dyes to Annexin V for targeted detection of apoptotic cells. Chem. Commun. 53, 368– 371, DOI: 10.1039/C6CC08671C39Site-selective installation of BASHY fluorescent dyes to Annexin V for targeted detection of apoptotic cellsCal, Pedro M. S. D.; Sieglitz, Florian; Santos, Fabio M. F.; Parente Carvalho, Catia; Guerreiro, Ana; Bertoldo, Jean B.; Pischel, Uwe; Gois, Pedro M. P.; Bernardes, Goncalo J. L.Chemical Communications (Cambridge, United Kingdom) (2017), 53 (2), 368-371CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)Fluorophores are indispensable for imaging biol. processes. The authors report the design and synthesis of azide-tagged boronic acid salicylidenehydrazone (BASHY) dyes and their use for site-selective labeling of Annexin V. The Annexin V-BASHY conjugate maintained function and fluorescence as demonstrated by the targeted detection of apoptotic cells.
- 40Ng, D. Y. W., Vill, R., Wu, Y., Koynov, K., Tokura, Y., Liu, W., Sihler, S., Kreyes, A., Ritz, S., Barth, H., Ziener, U., and Weil, T. (2017) Directing intracellular supramolecular assembly with N-heteroaromatic quaterthiophene analogues. Nat. Commun. 8, 1850, DOI: 10.1038/s41467-017-02020-240Directing intracellular supramolecular assembly with N-heteroaromatic quaterthiophene analoguesNg David Y W; Wu Yuzhou; Koynov Kaloian; Tokura Yu; Liu Weina; Weil Tanja; Vill Roman; Tokura Yu; Liu Weina; Sihler Susanne; Kreyes Andreas; Ziener Ulrich; Weil Tanja; Ritz Sandra; Barth HolgerNature communications (2017), 8 (1), 1850 ISSN:.Self-assembly in situ, where synthetic molecules are programmed to organize in a specific and complex environment i.e., within living cells, can be a unique strategy to influence cellular functions. Here we present a small series of rationally designed oligothiophene analogues that specifically target, locate and dynamically self-report their supramolecular behavior within the confinement of a cell. Through the recognition of the terminal alkyl substituent and the amphiphilic pyridine motif, we show that the cell provides different complementary pathways for self-assembly that can be traced easily with fluorescence microscopy as their molecular organization emits in distinct fluorescent bands. Importantly, the control and induction of both forms are achieved by time, temperature and the use of the intracellular transport inhibitor, bafilomycin A1. We showcase the importance of both intrinsic (cell) and extrinsic (stimulus) factors for self-organization and the potential of such a platform toward developing synthetic functional components within living cells.
- 41Najera, C., Baldo, B., and Yus, M. (1988) Regio- and Stereo-selective Synthesis of β-Sulphonyl-a,β- Unsaturated Carbonyl Compounds. J. Chem. Soc., Perkin Trans. 1 1029, DOI: 10.1039/P19880001029There is no corresponding record for this reference.
- 42Guez, V., Roux, P., Navon, A., and Goldberg, M. E. (2002) Role of individual disulfide bonds in hen lysozyme early folding steps. Protein Sci. 11, 1136– 51, DOI: 10.1110/ps.396010242Role of individual disulfide bonds in hen lysozyme early folding stepsGuez, Valerie; Roux, Pascale; Navon, Amiel; Goldberg, Michel E.Protein Science (2002), 11 (5), 1136-1151CODEN: PRCIEI; ISSN:0961-8368. (Cold Spring Harbor Laboratory Press)To probe the role of individual disulfide bonds in the folding kinetics of hen lysozyme, variants with 2 mutations, C30A/C115A, C64A/C80A, and C76A/C94A, were constructed. The corresponding proteins, each lacking one disulfide bond, were produced in Escherichia coli as inclusion bodies and solubilized, purified, and renatured/oxidized using original protocols. Their enzymic, spectral, and hydrodynamic characteristics confirmed that their conformations were very similar to that of native wild-type (WT) lysozyme. Stopped-flow studies on the renaturation of these guanidine-HCl-unfolded proteins with their 3 disulfide bonds intact showed that, for the 3 variants, the native far-UV ellipticity was regained in a burst phase within the 4-ms instrument dead-time. The transient overshoots of far-UV ellipticity and Trp residue fluorescence that follow the burst phase, as well as the kinetics of transient 8-anilino-1-naphthalene-sulfonic acid (ANS) binding, were diversely affected depending on the variant. Together with previous reports on the folding kinetics of WT lysozyme carboxymethylated on Cys-6 and Cys-127, detailed anal. of the kinetics showed that: (1) none of the disulfide bonds were indispensable for the rapid formation (<4 ms) of the native-like secondary structure; (2) the 2 intra-α-domain disulfides (Cys-6-Cys-127 and Cys-30-Cys-115) must be simultaneously present to generate the trapped intermediate responsible for the slow folding population obsd. in WT lysozyme; and (3) the intra-β-domain (Cys-64-Cys-80) and the inter-αβ-domains (Cys-76-Cys-94) disulfide bonds did not affect the kinetics of formation of the trapped intermediate but were involved in its stability.
- 43Springsteen, G. and Wang, B. (2002) A detailed examination of boronic acid–diol complexation. Tetrahedron 58, 5291– 5300, DOI: 10.1016/S0040-4020(02)00489-143A detailed examination of boronic acid-diol complexationSpringsteen, Greg; Wang, BingheTetrahedron (2002), 58 (26), 5291-5300CODEN: TETRAB; ISSN:0040-4020. (Elsevier Science Ltd.)Boronic acids bind with compds. contg. diol moieties with high affinity through reversible boronate formation. However, the conditions that foster tight binding between the diol and the boronic acid are not well understood. Also, due to the multiple ionic states of both the boronic acid and boronate ester, the equil. consts. reported in the literature have not always been strictly defined, and therefore there is a lack of comparability between the reported values. To address these issues, a method was developed for examg. boronate ester stability using the fluorescent reporter Alizarin Red S. and this system has been used to det. the binding consts. of a series of diols, and as a basis from which to derive a no. of relationships that correlate the various equil. consts. in the literature.
- 44Shugar, D. (1952) The measurement of lysozyme activity and the ultra-violet inactivation of lysozyme. Biochim. Biophys. Acta 8, 302– 9, DOI: 10.1016/0006-3002(52)90045-044Measurement of lysozyme activity and the ultraviolet inactivation of lysozymeShugar, DavidBiochimica et Biophysica Acta (1952), 8 (), 302-9CODEN: BBACAQ; ISSN:0006-3002.Lysozyme (I) activity is detd. by measuring the decrease with time of the optical d. of a suspension of Micrococcus lysodeikticus. The analysis is made at pH 7.1 (M/15 phosphate buffer) in 3 ml. vol. (optical d. 0.5 to 0.75) to which 0.02-0.05 cc. of enzyme soln. is added. The readings are made at 2820 A. over a 4.5 min. period. Inactivation by ultraviolet light is a first order reaction, k = 0.158 min.-1. The quantum yield for inactivation at 2537 A. is 0.024 over the pH range 3.6-12.0. No apparent liberation of peptides or amino acids was observed but some photo-oxidation took place.
- 45Davies, R. C. and Neuberger, A. (1969) Modification of lysine and arginine residues of lysozyme and the effect of enzymatic activity. Biochim. Biophys. Acta - Enzymol. 178, 306– 317, DOI: 10.1016/0005-2744(69)90398-245Modification of lysine and arginine residues of lysozyme and the effect on enzymic activityDavies, Richard C.; Neuberger, AlbertBiochimica et Biophysica Acta, Enzymology (1969), 178 (2), 306-17CODEN: BBEZAD; ISSN:0924-1086.Acetylation of all 6 lysine residues of lysozyme (EC 3.2.1.17) abolished lytic action towards cells of Micrococcus lysodeikticus in 0.05M phosphate buffer (pH 6.2) but did not affect cleavage of the tetramer of N-acetylglucosamine (GlcNAc)4 obtained from chitin. Acetylated lysozyme still lysed cells in solns. of low ionic strength at pH 6.2. Compared with unmodified enzyme the activity profile for acetyl lysozyme was displaced to much lower values of ionic strength and was still markedly dependent on pH. Chem. modification of the lysine groups with ethyl acetamidate increased the activity towards cells slightly, yet did not alter the activity towards (GlcNAc)4. Modification of 7 out of 8 arginine residues with 2,3-butanedione in borate buffer reduced the activity of acetyl lysozyme towards cells but not towards (GlcNAc)4. Since all the basic residues of lysozyme apparently lie outside the active center, the persistence of lytic activity over a very wide pH range is discussed in terms of the currently accepted mechanism of action of lysozyme.
- 46Imoto, T., Moriyama, S., and Yagishita, K. (1976) A Study of the Native-Denatured (N to/from D) Transition in Lysozyme III. Effect of Alteration of Net Charge by Acetylation. J. Biochem. 80, 1319– 1325, DOI: 10.1093/oxfordjournals.jbchem.a13140446A study of the native-denatured (N .dblharw. D) transition in lysozyme. III. Effect of alteration of net charge by acetylationImoto, Taiji; Moriyama, Sigeru; Yagishita, KazuyoshiJournal of Biochemistry (1976), 80 (6), 1319-25CODEN: JOBIAO; ISSN:0021-924X.Measurement of the enzymic activity and fluorescence properties showed that the gross conformation of acetylated lysozyme (I) is very similar to that of native I. On the other hand, protease digestion, t-Bu hypochloride modification, and thermal denaturation of native, acetylated, and guanidinated I showed that acetylation causes a small but significant shift of the N.dblharw.D transition to the right. Thus, charge balance in a protein plays an important role in maintaining its conformation. The difference between equil. and kinetic methods of monitoring protein denaturation was also clarified.
- 47Masuda, T., Kitabatake, N., and Ide, N. (2005) Effects of Chemical Modification of Lysine Residues on the Sweetness of Lysozyme. Chem. Chem. Senses 30, 253– 264, DOI: 10.1093/chemse/bji02147Effects of Chemical Modification of Lysine Residues on the Sweetness of LysozymeMasuda, Tetsuya; Ide, Nobuyuki; Kitabatake, NaofumiChemical Senses (2005), 30 (3), 253-264CODEN: CHSED8; ISSN:0379-864X. (Oxford University Press)Lysozyme is a sweet-tasting protein with a sweetness threshold value of around 7 μM. To clarify the effect of basicity at the side chain of lysine residues on the threshold values of sweetness, charge-specific chem. modifications such as guanidination, acetylation and phosphopyridoxylation of lysine residues were performed. Sensory anal. showed that the sweetness threshold value of lysozyme was not changed by guanidination, whereas it was increased markedly by acetylation and phosphopyridoxylation. To confirm the importance of the basicity in the lysine residues in detail, purifn. of acetylated (Ac-) and phosphopyridoxylated (PLP-) lysozymes using SP-ion exchange column chromatog. was performed. The threshold values were not changed by modification with fewer than two residues (∼7 μM), whereas the threshold values significantly increased to 15 and 34 μM when tetra-Ac and tri-PLP, resp. Furthermore, sweetness was not detected at 30 μM (hexa-, penta-Ac and tetra-PLP). It should be noted that removal of the neg. charges of the phosphate groups in the tri-PLP lysozyme by acid phosphatase resulted in the recovery of sweetness (6.4 μM), indicating that basicity at the position of the lysine residues is responsible for lysozyme sweetness and that strict charge complementarities might be required for interaction to its putative receptor.
- 48O’Brien, E. C., Farkas, E., Gil, M. J., Fitzgerald, D., Castineras, A., and Nolan, K. B. (2000) Metal complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid and benzohydroxamic acid. Crystal and molecular structure of [Cu(phen)2(Cl)]Cl·H2Sha, a model for a peroxidase-inhibitor complex. J. Inorg. Biochem. 79, 47– 51, DOI: 10.1016/S0162-0134(99)00245-748Metal complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid and benzohydroxamic acid. Crystal and molecular structure of [Cu(phen)2(Cl)]Cl·H2Sha, a model for a peroxidase-inhibitor complexO'Brien, Eimear C.; Farkas, Etelka; Gil, Marie Jose; Fitzgerald, Desmond; Castineras, Alfonso; Nolan, Kevin B.Journal of Inorganic Biochemistry (2000), 79 (1-4), 47-51CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier Science Inc.)Stability consts. of iron(III), copper(II), nickel(II) and zinc(II) complexes of salicylhydroxamic acid (H2Sha), anthranilic hydroxamic acid (HAha) and benzohydroxamic acid (HBha) have been detd. at 25.0°C, I=0.2 mol dm-3 KCl in aq. soln. The complex stability order, iron(III) » copper(II) > nickel(II) ∼ zinc(II) was obsd. while complexes of H2Sha were found to be more stable than those of the other two ligands. In the prepn. of ternary metal ion complexes of these ligands and 1,10-phenanthroline (phen) the cryst. complex [Cu(phen)2(Cl)]Cl·H2Sha was obtained and its crystal structure detd. This complex is a model for hydroxamate-peroxidase inhibitor interactions.
- 49Yang, S.-M., Lagu, B., and Wilson, L. J. (2007) Mild and Efficient Lewis Acid-Promoted Detritylation in the Synthesis of N -Hydroxy Amides: A Concise Synthesis of (−)-Cobactin T. J. Org. Chem. 72, 8123– 8126, DOI: 10.1021/jo701411dThere is no corresponding record for this reference.
- 50Shin, S. B. Y., Almeida, R. D., Gerona-Navarro, G., Bracken, C., and Jaffrey, S. R. (2010) Assembling ligands in situ using bioorthogonal boronate ester synthesis. Chem. Biol. 17, 1171– 6, DOI: 10.1016/j.chembiol.2010.09.00850Assembling ligands in situ using bioorthogonal boronate ester synthesisShin, Sung Bin Y.; Almeida, Ramiro D.; Gerona-Navarro, Guillermo; Bracken, Clay; Jaffrey, Samie R.Chemistry & Biology (Cambridge, MA, United States) (2010), 17 (11), 1171-1176CODEN: CBOLE2; ISSN:1074-5521. (Cell Press)Many mols. that could manipulate cellular function are not practical due to their large size and concomitant undesirable pharmacokinetic properties. Here, we describe a bioorthogonal, highly stable boronate ester (HiSBE) synthesis and use this reaction to synthesize a biol. active mol. from smaller precursors in a physiol. context. The rapid rate of HiSBE synthesis suggests that it may be useful for assembling a wide variety of biol. active mols. in physiol. solns.
- 51Shrestha, D., Jenei, A., Nagy, P., Vereb, G., and Szöllősi, J. (2015) Understanding FRET as a research tool for cellular studies. Int. J. Mol. Sci. 16, 6718– 56, DOI: 10.3390/ijms1604671851Understanding FRET as a research tool for cellular studiesShrestha, Dilip; Jenei, Attila; Nagy, Peter; Vereb, Gyoergy; Szoellosi, JanosInternational Journal of Molecular Sciences (2015), 16 (4), 6718-6756CODEN: IJMCFK; ISSN:1422-0067. (MDPI AG)Communication of mol. species through dynamic assocn. and/or dissocn. at various cellular sites governs biol. functions. Understanding these physiol. processes require delineation of mol. events occurring at the level of individual complexes in a living cell. Among the few non-invasive approaches with nanometer resoln. are methods based on Forster Resonance Energy Transfer (FRET). FRET is effective at a distance of 1-10 nm which is equiv. to the size of macromols., thus providing an unprecedented level of detail on mol. interactions. The emergence of fluorescent proteins and SNAP- and CLIP- tag proteins provided FRET with the capability to monitor changes in a mol. complex in real-time making it possible to establish the functional significance of the studied mols. in a native environment. Now, FRET is widely used in biol. sciences, including the field of proteomics, signal transduction, diagnostics and drug development to address questions almost unimaginable with biochem. methods and conventional microscopies. However, the underlying physics of FRET often scares biologists. Therefore, in this review, our goal is to introduce FRET to non-physicists in a lucid manner. We will also discuss our contributions to various FRET methodologies based on microscopy and flow cytometry, while describing its application for detg. the mol. heterogeneity of the plasma membrane in various cell types.
- 52Yano, Y. and Matsuzaki, K. (2009) Tag–probe labeling methods for live-cell imaging of membrane proteins. Biochim. Biophys. Acta, Biomembr. 1788, 2124– 2131, DOI: 10.1016/j.bbamem.2009.07.01752Tag-probe labeling methods for live-cell imaging of membrane proteinsYano, Yoshiaki; Matsuzaki, KatsumiBiochimica et Biophysica Acta, Biomembranes (2009), 1788 (10), 2124-2131CODEN: BBBMBS; ISSN:0005-2736. (Elsevier B.V.)A review. Instead of using reconstituted proteoliposomes, in situ investigations of membrane proteins in living cell membranes are important because the heterogeneous and dynamic nature of biomembranes significantly affects their behavior. Protein-specific labeling is a key technique for the detection of a target protein by fluorescence measurements, particularly fluorescence microscopy. However, conventional genetic fusion with fluorescent proteins has several shortcomings. Post-translational labeling methods using a genetically encodable tag and synthetic probes targeting to the tag can overcome these limitations. This review summarizes emerging tag-probe techniques for labeling specific membrane proteins and their applications, including endocytic internalization, partitioning to specific membrane domains, interprotein interactions, and conformational changes.
- 53Soriano-Ursúa, M. A., Farfán-García, E. D., López-Cabrera, Y., Querejeta, E., and Trujillo-Ferrara, J. G. (2014) Boron-containing acids: Preliminary evaluation of acute toxicity and access to the brain determined by Raman scattering spectroscopy. NeuroToxicology 40, 8– 15, DOI: 10.1016/j.neuro.2013.10.00553Boron-containing acids: Preliminary evaluation of acute toxicity and access to the brain determined by Raman scattering spectroscopySoriano-Ursua, Marvin A.; Farfan-Garcia, Eunice D.; Lopez-Cabrera, Yessica; Querejeta, Enrique; Trujillo-Ferrara, Jose G.NeuroToxicology (2014), 40 (), 8-15CODEN: NRTXDN; ISSN:0161-813X. (Elsevier Inc.)Boron-contg. compds. (BCCs), particularly boron contg. acids (BCAs), have become attractive moieties or mols. in drug development. It has been suggested that when functional groups with boron atoms are added to well-known drugs, the latter are conferred with greater potency and efficacy in relation to their target receptors. However, the use of BCAs in drug development is limited due to the lack of a toxicol. profile. Consequently, the aim of the present study was to evaluate the acute toxicity of boric and boronic acids. Thus, a detn. was made of the LD (LD50) of test compds. in male CD1 mice, as well as the ED required to neg. affect spontaneous motor activity and to produce notable behavioral abnormalities. After treatment of animals at different doses, macroscopic observations were made from a necropsy, and Raman scattering spectroscopic studies were carried out on brain tissue samples. In general, the results show that most of the tested BCAs have very low toxicity, evidenced by the high doses required to induce notable toxic effects (greater than 100 mg/kg of body wt. for all compds., except for 3-thyenilboronic acid). Such toxic effects, presumably mediated by action on the CNS, include eye damage, gastrointestinal effects (e.g., gastric-gut dilatation and fecal retention), sedation, hypnosis and/or trembling. This preliminary toxicol. profile suggests that BCAs can be considered potential therapeutic agents or moieties to be added to other compds. in the development of new drugs. Future studies are required to explore possible chronic toxicity of BCCs.
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.8b00358.
Full experimental procedures and characterization data for new compounds (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.