Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Biomacromolecules ; 25(5): 3001-3010, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38598264

ABSTRACT

Glycosylation is a valuable tool for modulating protein solubility; however, the lack of reliable research strategies has impeded efficient progress in understanding and applying this modification. This study aimed to bridge this gap by investigating the solubility of a model glycoprotein molecule, the carbohydrate-binding module (CBM), through a two-stage process. In the first stage, an approach involving chemical synthesis, comparative analysis, and molecular dynamics simulations of a library of glycoforms was employed to elucidate the effect of different glycosylation patterns on solubility and the key factors responsible for the effect. In the second stage, a predictive mathematical formula, innovatively harnessing machine learning algorithms, was derived to relate solubility to the identified key factors and accurately predict the solubility of the newly designed glycoforms. Demonstrating feasibility and effectiveness, this two-stage approach offers a valuable strategy for advancing glycosylation research, especially for the discovery of glycoforms with increased solubility.


Subject(s)
Machine Learning , Molecular Dynamics Simulation , Solubility , Glycosylation , Glycoproteins/chemistry
2.
J Comput Aided Mol Des ; 36(4): 313-328, 2022 04.
Article in English | MEDLINE | ID: mdl-35507105

ABSTRACT

Insulin has been commonly adopted as a peptide drug to treat diabetes as it facilitates the uptake of glucose from the blood. The development of oral insulin remains elusive over decades owing to its susceptibility to the enzymes in the gastrointestinal tract and poor permeability through the intestinal epithelium upon dimerization. Recent experimental studies have revealed that certain O-linked glycosylation patterns could enhance insulin's proteolytic stability and reduce its dimerization propensity, but understanding such phenomena at the molecular level is still difficult. To address this challenge, we proposed and tested several structural determinants that could potentially influence insulin's proteolytic stability and dimerization propensity. We used these metrics to assess the properties of interest from [Formula: see text] aggregate molecular dynamics of each of 12 targeted insulin glyco-variants from multiple wild-type crystal structures. We found that glycan-involved hydrogen bonds and glycan-dimer occlusion were useful metrics predicting the proteolytic stability and dimerization propensity of insulin, respectively, as was in part the solvent-accessible surface area of proteolytic sites. However, other plausible metrics were not generally predictive. This work helps better explain how O-linked glycosylation influences the proteolytic stability and monomeric propensity of insulin, illuminating a path towards rational molecular design of insulin glycoforms.


Subject(s)
Insulin , Molecular Dynamics Simulation , Dimerization , Insulin/analogs & derivatives , Insulin/chemistry , Polysaccharides
3.
Molecules ; 27(24)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36557993

ABSTRACT

Therapeutic proteins have unique advantages over small-molecule drugs in the treatment of various diseases, such as higher target specificity, stronger pharmacological efficacy and relatively low side effects. These advantages make them increasingly valued in drug development and clinical practice. However, although highly valued, the intrinsic limitations in their physical, chemical and pharmacological properties often restrict their wider applications. As one of the most important post-translational modifications, glycosylation has been shown to exert positive effects on many properties of proteins, including molecular stability, and pharmacodynamic and pharmacokinetic characteristics. Glycoengineering, which involves changing the glycosylation patterns of proteins, is therefore expected to be an effective means of overcoming the problems of therapeutic proteins. In this review, we summarize recent efforts and advances in the glycoengineering of erythropoietin and IgG monoclonal antibodies, with the goals of illustrating the importance of this strategy in improving the performance of therapeutic proteins and providing a brief overview of how glycoengineering is applied to protein-based drugs.


Subject(s)
Antibodies, Monoclonal , Protein Engineering , Glycosylation , Antibodies, Monoclonal/metabolism , Protein Processing, Post-Translational , Immunoglobulin G/chemistry , Polysaccharides/metabolism
4.
Biochemistry ; 60(46): 3455-3469, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34196546

ABSTRACT

Since the establishment of site-specific mutagenesis of single amino acids to interrogate protein function in the 1970s, biochemists have sought to tailor protein structure in the native cell environment. Fine-tuning the chemical properties of proteins is an indispensable way to address fundamental mechanistic questions. Unnatural amino acids (UAAs) offer the possibility to expand beyond the 20 naturally occurring amino acids in most species and install new and useful chemical functions. Here, we review the literature about advances in UAA incorporation technology from chemoenzymatic aminoacylation of modified tRNAs to in vitro translation systems to genetic encoding of UAAs in the native cell environment and whole organisms. We discuss innovative applications of the UAA technology to challenges in bioengineering and medicine.


Subject(s)
Biochemistry/history , Genetic Code , Amino Acids/genetics , Amino Acids/metabolism , Biochemistry/methods , Biochemistry/trends , History, 20th Century , History, 21st Century , Mutagenesis, Site-Directed , RNA, Transfer/genetics , RNA, Transfer/metabolism
5.
J Biol Chem ; 295(21): 7341-7349, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32295844

ABSTRACT

The role of O-linked N-acetylglucosamine (O-GlcNAc) modification in the cell cycle has been enigmatic. Previously, both O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) disruptions have been shown to derail the mitotic centrosome numbers, suggesting that mitotic O-GlcNAc oscillation needs to be in concert with mitotic progression to account for centrosome integrity. Here, using both chemical approaches and biological assays with HeLa cells, we attempted to address the underlying molecular mechanism and observed that incubation of the cells with the OGA inhibitor Thiamet-G strikingly elevates centrosomal distances, suggestive of premature centrosome disjunction. These aberrations could be overcome by inhibiting Polo-like kinase 1 (PLK1), a mitotic master kinase. PLK1 inactivation is modulated by the myosin phosphatase targeting subunit 1 (MYPT1)-protein phosphatase 1cß (PP1cß) complex. Interestingly, MYPT1 has been shown to be abundantly O-GlcNAcylated, and the modified residues have been detected in a recent O-GlcNAc-profiling screen utilizing chemoenzymatic labeling and bioorthogonal conjugation. We demonstrate here that MYPT1 is O-GlcNAcylated at Thr-577, Ser-585, Ser-589, and Ser-601, which antagonizes CDK1-dependent phosphorylation at Ser-473 and attenuates the association between MYPT1 and PLK1, thereby promoting PLK1 activity. We conclude that under high O-GlcNAc levels, PLK1 is untimely activated, conducive to inopportune centrosome separation and disruption of the cell cycle. We propose that too much O-GlcNAc is equally deleterious as too little O-GlcNAc, and a fine balance between the OGT/OGA duo is indispensable for successful mitotic divisions.


Subject(s)
Centrosome/metabolism , Mitosis , Myosin-Light-Chain Phosphatase/metabolism , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Glycosylation , Humans , Myosin-Light-Chain Phosphatase/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
6.
Proc Natl Acad Sci U S A ; 114(52): 13667-13672, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229855

ABSTRACT

In nature, many microbes secrete mixtures of glycoside hydrolases, oxidoreductases, and accessory enzymes to deconstruct polysaccharides and lignin in plants. These enzymes are often decorated with N- and O-glycosylation, the roles of which have been broadly attributed to protection from proteolysis, as the extracellular milieu is an aggressive environment. Glycosylation has been shown to sometimes affect activity, but these effects are not fully understood. Here, we examine N- and O-glycosylation on a model, multimodular glycoside hydrolase family 7 cellobiohydrolase (Cel7A), which exhibits an O-glycosylated carbohydrate-binding module (CBM) and an O-glycosylated linker connected to an N- and O-glycosylated catalytic domain (CD)-a domain architecture common to many biomass-degrading enzymes. We report consensus maps for Cel7A glycosylation that include glycan sites and motifs. Additionally, we examine the roles of glycans on activity, substrate binding, and thermal and proteolytic stability. N-glycan knockouts on the CD demonstrate that N-glycosylation has little impact on cellulose conversion or binding, but does have major stability impacts. O-glycans on the CBM have little impact on binding, proteolysis, or activity in the whole-enzyme context. However, linker O-glycans greatly impact cellulose conversion via their contribution to proteolysis resistance. Molecular simulations predict an additional role for linker O-glycans, namely that they are responsible for maintaining separation between ordered domains when Cel7A is engaged on cellulose, as models predict α-helix formation and decreased cellulose interaction for the nonglycosylated linker. Overall, this study reveals key roles for N- and O-glycosylation that are likely broadly applicable to other plant cell-wall-degrading enzymes.


Subject(s)
Cellulase/metabolism , Polysaccharides/metabolism , Cellulase/chemistry , Enzyme Activation , Enzyme Stability , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , Glycosylation , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Models, Molecular , Molecular Conformation , Polysaccharides/chemistry , Proteolysis , Transition Temperature
7.
Biochemistry ; 57(4): 413-428, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29309128

ABSTRACT

Protein glycosylation is one of the most common post-translational modifications and can influence many properties of proteins. Abnormal protein glycosylation can lead to protein malfunction and serious disease. While appreciation of glycosylation's importance is growing in the scientific community, especially in recent years, a lack of homogeneous glycoproteins with well-defined glycan structures has made it difficult to understand the correlation between the structure of glycoproteins and their properties at a quantitative level. This has been a significant limitation on rational applications of glycosylation and on optimizing glycoprotein properties. Through the extraordinary efforts of chemists, it is now feasible to use chemical synthesis to produce collections of homogeneous glycoforms with systematic variations in amino acid sequence, glycosidic linkage, anomeric configuration, and glycan structure. Such a technical advance has greatly facilitated the study and application of protein glycosylation. This Perspective highlights some representative work in this research area, with the goal of inspiring and encouraging more scientists to pursue the glycosciences.


Subject(s)
Glycoproteins/chemical synthesis , Protein Engineering/methods , Protein Processing, Post-Translational , Amino Acid Sequence , Forecasting , Glycopeptides/chemical synthesis , Glycosylation , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutagenesis, Site-Directed , Polysaccharides/chemistry , Protein Conformation , Protein Stability
8.
Biochemistry ; 57(1): 136-148, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29202246

ABSTRACT

Many human proteins have the potential to be developed as therapeutic agents. However, side effects caused by direct administration of natural proteins have significantly slowed expansion of protein therapeutics into the clinic. Post-translational modifications (PTMs) can improve protein properties, but because of significant knowledge gaps, we are considerably limited in our ability to apply PTMs to generate better protein therapeutics. Here, we seek to fill the gaps by studying the PTMs of a small representative chemotactic cytokine, RANTES. RANTES can inhibit HIV-1 infection by competing with it for binding to receptor CCR5 and stimulating CCR5 endocytosis. Unfortunately, RANTES can induce strong signaling, leading to severe inflammatory side effects. We apply a chemical biology approach to explore the potential of post-translationally modified RANTES as safe inhibitors of HIV-1 infection. We synthesized and systematically tested a library of RANTES isoforms for their ability to inhibit inflammatory signaling and prevent HIV-1 infection of primary human cells. Through this research, we revealed that most of the glycosylated variants have decreased inflammation-associated properties and identified one particular glyco variant, a truncated RANTES containing a Galß1-3GalNAc disaccharide α-linked to Ser4, which stands out as having the best overall properties: relatively high HIV-1 inhibition potency but also weak inflammatory properties. Moreover, our results provided a structural basis for the observed changes in the properties of RANTES. Taken together, this work highlights the potential importance of glycosylation as an alternative strategy for developing CCR5 inhibitors to treat HIV-1 infection and, more generally, for reducing or eliminating unwanted properties of therapeutic proteins.


Subject(s)
Chemokine CCL5/chemistry , Chemokine CCL5/pharmacology , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Protein Processing, Post-Translational , Acylation , Biopolymers , Carbon-13 Magnetic Resonance Spectroscopy , Chemokine CCL5/adverse effects , Chemokine CCL5/metabolism , Chemotaxis, Leukocyte/drug effects , Glycosaminoglycans/metabolism , Glycosylation , HIV Fusion Inhibitors/adverse effects , HIV Fusion Inhibitors/metabolism , HIV Infections/drug therapy , HIV-1/physiology , Humans , Proton Magnetic Resonance Spectroscopy , Receptors, CCR5/metabolism , THP-1 Cells
9.
Proteins ; 86(2): 164-176, 2018 02.
Article in English | MEDLINE | ID: mdl-29127727

ABSTRACT

SH2B1 is a multidomain protein that serves as a key adaptor to regulate numerous cellular events, such as insulin, leptin, and growth hormone signaling pathways. Many of these protein-protein interactions are mediated by the SH2 domain of SH2B1, which recognizes ligands containing a phosphorylated tyrosine (pY), including peptides derived from janus kinase 2, insulin receptor, and insulin receptor substrate-1 and -2. Specificity for the SH2 domain of SH2B1 is conferred in these ligands either by a hydrophobic or an acidic side chain at the +3 position C-terminal to the pY. This specificity for chemically disparate species suggests that SH2B1 relies on distinct thermodynamic or structural mechanisms to bind to peptides. Using binding and structural strategies, we have identified unique thermodynamic signatures for each peptide binding mode, and several SH2B1 residues, including K575 and R578, that play distinct roles in peptide binding. The high-resolution structure of the SH2 domain of SH2B1 further reveals conformationally plastic protein loops that may contribute to the ability of the protein to recognize dissimilar ligands. Together, numerous hydrophobic and electrostatic interactions, in addition to backbone conformational flexibility, permit the recognition of diverse peptides by SH2B1. An understanding of this expanded peptide recognition will allow for the identification of novel physiologically relevant SH2B1/peptide interactions, which can contribute to the design of obesity and diabetes pharmaceuticals to target the ligand-binding interface of SH2B1 with high specificity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Peptides/metabolism , src Homology Domains , Adaptor Proteins, Signal Transducing/chemistry , Crystallography, X-Ray , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Interaction Maps , Static Electricity , Thermodynamics
10.
Biochemistry ; 56(16): 2225-2237, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28376302

ABSTRACT

SH2 domains recognize phosphotyrosine (pY)-containing peptide ligands and play key roles in the regulation of receptor tyrosine kinase pathways. Each SH2 domain has individualized specificity, encoded in the amino acids neighboring the pY, for defined targets that convey their distinct functions. The C-terminal SH2 domain (PLCC) of the phospholipase C-γ1 full-length protein (PLCγ1) typically binds peptides containing small and hydrophobic amino acids adjacent to the pY, including a peptide derived from platelet-derived growth factor receptor B (PDGFRB) and an intraprotein recognition site (Y783 of PLCγ1) involved in the regulation of the protein's lipase activity. Remarkably, PLCC also recognizes unexpected peptides containing amino acids with polar or bulky side chains that deviate from this pattern. This versatility in recognition specificity may allow PLCγ1 to participate in diverse, previously unrecognized, signaling pathways in response to binding chemically dissimilar partners. We have used structural approaches, including nuclear magnetic resonance and X-ray crystallography, to elucidate the mechanisms of noncognate peptide binding to PLCC by ligands derived from receptor tyrosine kinase ErbB2 and from the insulin receptor. The high-resolution peptide-bound structures reveal that PLCC has a relatively static backbone but contains a chemically rich protein surface comprised of a combination of hydrophobic pockets and amino acids with charged side chains. We demonstrate that this expansive and chemically diverse PLCC interface, in addition to peptide conformational plasticity, permits PLCC to recognize specific noncognate peptide ligands with multimodal specificity.


Subject(s)
Peptides/metabolism , Phospholipase C gamma/metabolism , src Homology Domains , Binding Sites , Calorimetry , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptides/chemistry , Phospholipase C gamma/chemistry , Protein Conformation
11.
Biochemistry ; 56(23): 2897-2906, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28494147

ABSTRACT

Protein glycosylation has been shown to have a variety of site-specific and glycan-specific effects, but so far, the molecular logic that leads to such observations has been elusive. Understanding the structural changes that occur and being able to correlate those with the physical properties of the glycopeptide are valuable steps toward being able to predict how specific glycosylation patterns will affect the stability of glycoproteins. By systematically comparing the structural features of the O-glycosylated carbohydrate-binding module of a Trichoderma reesei-derived Family 7 cellobiohydrolase, we were able to develop a better understanding of the influence of O-glycan structure on the molecule's physical stability. Our results indicate that the previously observed stabilizing effects of O-glycans come from the introduction of new bonding interactions to the structure and increased rigidity, while the decreased stability seemed to result from the impaired interactions and increased conformational flexibility. This type of knowledge provides a powerful and potentially general mechanism for improving the stability of proteins through glycoengineering.


Subject(s)
Cellulose 1,4-beta-Cellobiosidase/metabolism , Fungal Proteins/metabolism , Glycoproteins/metabolism , Glycoside Hydrolases/metabolism , Models, Molecular , Protein Processing, Post-Translational , Trichoderma/enzymology , Amino Acid Substitution , Binding Sites , Cellulose 1,4-beta-Cellobiosidase/chemistry , Enzyme Stability , Fungal Proteins/chemistry , Glycoproteins/chemistry , Glycoside Hydrolases/chemistry , Glycosylation , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Folding , Protein Unfolding
12.
Biochemistry ; 56(34): 4539-4548, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28745859

ABSTRACT

Protein O-glycosylation is a diverse, common, and important post-translational modification of both proteins inside the cell and those that are secreted or membrane-bound. Much work has shown that O-glycosylation can alter the structure, function, and physical properties of the proteins to which it is attached. One gap remaining in our understanding of O-glycoproteins is how O-glycans might affect the folding of proteins. Here, we took advantage of synthetic, homogeneous O-glycopeptides to show that certain glycosylation patterns have an intrinsic effect, independent of any cellular folding machinery, on the folding pathway of a model O-glycoprotein, a carbohydrate binding module (CBM) derived from the Trichoderma reesei cellulase TrCel7A. The strongest effect, a 6-fold increase in overall folding rate, was observed when a single O-mannose was the glycan, and the glycosylation site was near the N-terminus of the peptide sequence. We were also able to show that glycosylation patterns affected the kinetics of each step in unique ways, which may help to explain the observations made here. This work is a first step toward quantitative understanding of how O-glycosylation might control, through intrinsic means, the folding of O-glycoproteins. Such an understanding is expected to facilitate future investigations into the effects of glycosylation on more biological processes related to protein folding.


Subject(s)
Cellulase/metabolism , Fungal Proteins/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Protein Folding , Trichoderma/enzymology , Cellulase/chemistry , Cellulase/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Glycoproteins/chemistry , Glycoproteins/genetics , Polysaccharides/chemistry , Polysaccharides/genetics , Trichoderma/genetics
13.
Proc Natl Acad Sci U S A ; 111(21): 7612-7, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821760

ABSTRACT

The majority of biological turnover of lignocellulosic biomass in nature is conducted by fungi, which commonly use Family 1 carbohydrate-binding modules (CBMs) for targeting enzymes to cellulose. Family 1 CBMs are glycosylated, but the effects of glycosylation on CBM function remain unknown. Here, the effects of O-mannosylation are examined on the Family 1 CBM from the Trichoderma reesei Family 7 cellobiohydrolase at three glycosylation sites. To enable this work, a procedure to synthesize glycosylated Family 1 CBMs was developed. Subsequently, a library of 20 CBMs was synthesized with mono-, di-, or trisaccharides at each site for comparison of binding affinity, proteolytic stability, and thermostability. The results show that, although CBM mannosylation does not induce major conformational changes, it can increase the thermolysin cleavage resistance up to 50-fold depending on the number of mannose units on the CBM and the attachment site. O-Mannosylation also increases the thermostability of CBM glycoforms up to 16 °C, and a mannose disaccharide at Ser3 seems to have the largest themostabilizing effect. Interestingly, the glycoforms with small glycans at each site displayed higher binding affinities for crystalline cellulose, and the glycoform with a single mannose at each of three positions conferred the highest affinity enhancement of 7.4-fold. Overall, by combining chemical glycoprotein synthesis and functional studies, we show that specific glycosylation events confer multiple beneficial properties on Family 1 CBMs.


Subject(s)
Biofuels , Cellulases/metabolism , Lignin/metabolism , Models, Molecular , Receptors, Cell Surface/metabolism , Trichoderma/enzymology , Cellulases/chemistry , Glycosylation , Lignin/chemistry , Molecular Structure , Protein Engineering/methods , Receptors, Cell Surface/chemistry
14.
Proc Natl Acad Sci U S A ; 110(36): 14646-51, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959893

ABSTRACT

Plant cell-wall polysaccharides represent a vast source of food in nature. To depolymerize polysaccharides to soluble sugars, many organisms use multifunctional enzyme mixtures consisting of glycoside hydrolases, lytic polysaccharide mono-oxygenases, polysaccharide lyases, and carbohydrate esterases, as well as accessory, redox-active enzymes for lignin depolymerization. Many of these enzymes that degrade lignocellulose are multimodular with carbohydrate-binding modules (CBMs) and catalytic domains connected by flexible, glycosylated linkers. These linkers have long been thought to simply serve as a tether between structured domains or to act in an inchworm-like fashion during catalytic action. To examine linker function, we performed molecular dynamics (MD) simulations of the Trichoderma reesei Family 6 and Family 7 cellobiohydrolases (TrCel6A and TrCel7A, respectively) bound to cellulose. During these simulations, the glycosylated linkers bind directly to cellulose, suggesting a previously unknown role in enzyme action. The prediction from the MD simulations was examined experimentally by measuring the binding affinity of the Cel7A CBM and the natively glycosylated Cel7A CBM-linker. On crystalline cellulose, the glycosylated linker enhances the binding affinity over the CBM alone by an order of magnitude. The MD simulations before and after binding of the linker also suggest that the bound linker may affect enzyme action due to significant damping in the enzyme fluctuations. Together, these results suggest that glycosylated linkers in carbohydrate-active enzymes, which are intrinsically disordered proteins in solution, aid in dynamic binding during the enzymatic deconstruction of plant cell walls.


Subject(s)
Cellulose 1,4-beta-Cellobiosidase/metabolism , Cellulose/metabolism , Fungal Proteins/metabolism , Lignin/metabolism , Binding Sites , Binding, Competitive , Biocatalysis , Catalytic Domain , Cellulose 1,4-beta-Cellobiosidase/chemistry , Fungal Proteins/chemistry , Glycosylation , Hydrolysis , Mass Spectrometry , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Trichoderma/enzymology , Trichoderma/metabolism
15.
Top Curr Chem ; 363: 155-92, 2015.
Article in English | MEDLINE | ID: mdl-25707614

ABSTRACT

Chemical protein synthesis is a useful tool to generate pure proteins which are otherwise difficult to obtain in sufficient amounts for structure and property analysis. Additionally, because of the precise and flexible nature of chemical synthesis, it allows for controllable variation of protein sequences, which is valuable for understanding the relationships between protein structure and function. Despite the usefulness of chemical protein synthesis, it has not been widely adopted as a tool for protein characterization, mainly because of the lack of general and efficient methods for the preparation and coupling of peptide fragments and for the folding of polypeptide chains. To address these issues, many new methods have recently been developed in the areas of solid-phase peptide synthesis, peptide fragment assembly, and protein folding. Here we review these recent technological advances and highlight the gaps needing to be addressed in future research.


Subject(s)
Chemistry Techniques, Synthetic/methods , Protein Engineering/methods , Proteins/chemical synthesis , Models, Molecular , Protein Conformation
16.
Proc Natl Acad Sci U S A ; 108(11): 4297-302, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368182

ABSTRACT

The conversion of peptide and proteins from their soluble state into well-organized aggregates, together with the accompanied oxidation of methionine residue, presents a significant challenge to human health, to the manufacture of protein therapeutics, and to the synthesis of proteins and glycoproteins. Despite their fundamental importance, little is known about the molecular basis of these two side reactions and their control. Here, using chemical peptide synthesis, we further confirmed the importance of the balance between hydrophobic interactions and electrostatic repulsive forces in inducing and inhibiting aggregation and methionine oxidation. Most importantly, through extending the established principle, we are able to effectively stabilize the problematic peptide fragment through the attachment of cleavable arginine tags. Future applications of our approach are expected to facilitate the synthesis and study of difficult peptides, proteins, and glycoproteins and will provide more opportunities for the optimization of protein biopharmaceuticals and for the development of cell-permeable biomolecules.


Subject(s)
Biochemistry/methods , Proteins/chemistry , Amino Acid Sequence , Chromatography, Liquid , Erythropoietin/chemistry , Humans , Mass Spectrometry , Methionine/metabolism , Molecular Sequence Data , Oxidation-Reduction , Peptides/chemistry , Protein Structure, Quaternary
17.
Proc Natl Acad Sci U S A ; 108(15): 5986-9, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21444787

ABSTRACT

The power of chemical synthesis of large cysteine-free polypeptides has been significantly enhanced through the use of nonproteogenic constructs which bear strategically placed thiol groups, enabling native chemical ligation. Central to these much expanded capabilities is the specific, radical-induced, metal-free dethiolation, which can be accomplished in aqueous medium.


Subject(s)
Cysteine/chemistry , Parathyroid Hormone/chemical synthesis , Amino Acid Sequence , Amino Acid Substitution , Circular Dichroism , Humans , Molecular Sequence Data , Parathyroid Hormone/chemistry
18.
Int J Biol Macromol ; 253(Pt 1): 126649, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37666405

ABSTRACT

There is an increasing interest in using S-glycosylation as a replacement for the more commonly occurring O-glycosylation, aiming to enhance the resistance of glycans against chemical hydrolysis and enzymatic degradation. However, previous studies have demonstrated that these two types of glycosylation exert distinct effects on protein properties and functions. In order to elucidate the structural basis behind the observed differences, we conducted a systematic and comparative analysis of 6 differently glycosylated forms of a model glycoprotein, CBM, using NMR spectroscopy and molecular dynamic simulations. Our findings revealed that the different stabilizing effects of S- and O-glycosylation could be attributed to altered hydrogen-bonding capability between the glycan and the polypeptide chain, and their diverse impacts on binding affinity could be elucidated by examining the interactions and motion dynamics of glycans in substrate-bound states. Overall, this study underscores the pivotal role of the glycosidic linkage in shaping the function of glycosylation and advises caution when switching glycosylation types in protein glycoengineering.


Subject(s)
Glycoproteins , Polysaccharides , Glycosylation , Glycoproteins/chemistry , Polysaccharides/metabolism , Peptides/chemistry , Magnetic Resonance Spectroscopy
19.
Int J Biol Macromol ; 235: 123833, 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-36870654

ABSTRACT

The role of glycosylation in the binding of glycoproteins to carbohydrate substrates has not been well understood. The present study addresses this knowledge gap by elucidating the links between the glycosylation patterns of a model glycoprotein, a Family 1 carbohydrate-binding module (TrCBM1), and the thermodynamic and structural properties of its binding to different carbohydrate substrates using isothermal titration calorimetry and computational simulation. The variations in glycosylation patterns cause a gradual transition of the binding to soluble cellohexaose from an entropy-driven process to an enthalpy-driven one, a trend closely correlated with the glycan-induced shift of the predominant binding force from hydrophobic interactions to hydrogen bonding. However, when binding to a large surface of solid cellulose, glycans on TrCBM1 have a more dispersed distribution and thus have less adverse impact on the hydrophobic interaction forces, leading to overall improved binding. Unexpectedly, our simulation results also suggest an evolutionary role of O-mannosylation in transforming the substrate binding features of TrCBM1 from those of type A CBMs to those of type B CBMs. Taken together, these findings provide new fundamental insights into the molecular basis of the role of glycosylation in protein-carbohydrate interactions and are expected to better facilitate further studies in this area.


Subject(s)
Cellulose , Polysaccharides , Glycosylation , Cellulose/chemistry , Computer Simulation , Thermodynamics , Protein Binding , Binding Sites
20.
J Am Chem Soc ; 134(8): 3912-6, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22332757

ABSTRACT

Application of native chemical ligation logic to the case of an N-terminal proline is described. Two approaches were studied. One involved incorporation of a 3R-substituted thiyl-proline derivative. Improved results were obtained from a 3R-substituted selenol function, incorporated in the context of an oxidized dimer.


Subject(s)
Proline/chemistry , Erythropoietin/chemical synthesis , Erythropoietin/chemistry , Molecular Structure , Proline/analogs & derivatives , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL