Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 10(23): e2301340, 2023 08.
Article in English | MEDLINE | ID: mdl-37290045

ABSTRACT

The treatment of implant-associated bacterial infections and biofilms is an urgent medical need and a grand challenge because biofilms protect bacteria from the immune system and harbor antibiotic-tolerant persister cells. This need is addressed herein through an engineering of antibody-drug conjugates (ADCs) that contain an anti-neoplastic drug mitomycin C, which is also a potent antimicrobial against biofilms. The ADCs designed herein release the conjugated drug without cell entry, via a novel mechanism of drug release which likely involves an interaction of ADC with the thiols on the bacterial cell surface. ADCs targeted toward bacteria are superior by the afforded antimicrobial effects compared to the non-specific counterpart, in suspension and within biofilms, in vitro, and in an implant-associated murine osteomyelitis model in vivo. The results are important in developing ADC for a new area of application with a significant translational potential, and in addressing an urgent medical need of designing a treatment of bacterial biofilms.


Subject(s)
Anti-Infective Agents , Immunoconjugates , Mice , Animals , Drug Liberation , Bacteria , Biofilms
2.
RSC Med Chem ; 14(6): 1186-1191, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37360394

ABSTRACT

The caseinolytic protease complex ClpXP is an important house-keeping enzyme in prokaryotes charged with the removal and degradation of misfolded and aggregated proteins and performing regulatory proteolysis. Dysregulation of its function, particularly by inhibition or allosteric activation of the proteolytic core ClpP, has proven to be a promising strategy to reduce virulence and eradicate persistent bacterial infections. Here, we report a rational drug-design approach to identify macrocyclic peptides which increase proteolysis by ClpP. This work expands the understanding of ClpP dynamics and sheds light on the conformational control exerted by its binding partner, the chaperone ClpX, by means of a chemical approach. The identified macrocyclic peptide ligands may, in the future, serve as a starting point for the development of ClpP activators for antibacterial applications.

3.
PLoS One ; 17(9): e0274606, 2022.
Article in English | MEDLINE | ID: mdl-36121820

ABSTRACT

One of the most straightforward and commonly used chemical modifications of proteins is to react surface amino groups (lysine residues) with activated esters. This chemistry has been used to generate protein-polymer conjugates, many of which are now approved therapeutics. Similar conjugates have also been generated by reacting activated ester atom transfer polymerization initiators with lysine residues to create biomacromolecular initiators for polymerization reactions. The reaction between activated esters and lysine amino groups is rapid and has been consistently described in almost every publication on the topic as a "random reaction". A random reaction implies that every accessible lysine amino group on a protein molecule is equally reactive, and as a result, that the reaction is indiscriminate. Nonetheless, the literature contradicts itself by also suggesting that some lysine amino groups are more reactive than others (as a function of pKa, surface accessibility, temperature, and local environment). If the latter assumption is correct, then the outcome of these reactions cannot be random at all, and we should be able to predict the outcome from the structure of the protein. Predicting the non-random outcome of a reaction between surface lysines and reactive esters could transform the speed at which active bioconjugates can be developed and engineered. Herein, we describe a robust integrated tool that predicts the activated ester reactivity of every lysine in a protein, thereby allowing us to calculate the non-random sequence of reaction as a function of reaction conditions. Specifically, we have predicted the intrinsic reactivity of each lysine in multiple proteins with a bromine-functionalised N-hydroxysuccinimide initiator molecule. We have also shown that the model applied to PEGylation. The rules-based analysis has been coupled together in a single Python program that can bypass tedious trial and error experiments usually needed in protein-polymer conjugate design and synthesis.


Subject(s)
Bromine , Lysine , Esters , Lysine/metabolism , Polymers/chemistry , Protein Processing, Post-Translational , Proteins/metabolism
4.
Bioconjug Chem ; 33(9): 1643-1653, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-35994522

ABSTRACT

PEGylation is a well-established and clinically proven half-life extension strategy for protein delivery. Protein modification with amine-reactive poly(ethylene glycol) (PEG) generates heterogeneous and complex bioconjugate mixtures, often composed of several PEG positional isomers with varied therapeutic efficacy. Laborious and costly experiments for reaction optimization and purification are needed to generate a therapeutically useful PEG conjugate. Kinetic models which accurately predict the outcome of so-called "random" PEGylation reactions provide an opportunity to bypass extensive wet lab experimentation and streamline the bioconjugation process. In this study, we propose a protein tertiary structure-dependent reactivity model that describes the rate of protein-amine PEGylation and introduces "PEG chain coverage" as a tangible metric to assess the shielding effect of PEG chains. This structure-dependent reactivity model was implemented into three models (linear, structure-based, and machine-learned) to gain insight into how protein-specific molecular descriptors (exposed surface areas, pKa, and surface charge) impacted amine reactivity at each site. Linear and machine-learned models demonstrated over 75% prediction accuracy with butylcholinesterase. Model validation with Somavert, PEGASYS, and phenylalanine ammonia lyase showed good correlation between predicted and experimentally determined degrees of modification. Our structure-dependent reactivity model was also able to simulate PEGylation progress curves and estimate "PEGmer" distribution with accurate predictions across different proteins, PEG linker chemistry, and PEG molecular weights. Moreover, in-depth analysis of these simulated reaction curves highlighted possible PEG conformational transitions (from dumbbell to brush) on the surface of lysozyme, as a function of PEG molecular weight.


Subject(s)
Data Science , Muramidase , Amines , Muramidase/chemistry , Phenylalanine Ammonia-Lyase , Polyethylene Glycols/chemistry , Proteins/chemistry
5.
Nat Commun ; 13(1): 4861, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35982075

ABSTRACT

We present three classes of chemical zymogens established around the protein cysteinome. In each case, the cysteine thiol group was converted into a mixed disulfide: with a small molecule, a non-degradable polymer, or with a fast-depolymerizing fuse polymer (ZLA). The latter was a polydisulfide based on naturally occurring molecule, lipoic acid. Zymogen designs were applied to cysteine proteases and a kinase. In each case, enzymatic activity was successfully masked in full and reactivated by small molecule reducing agents. However, only ZLA could be reactivated by protein activators, demonstrating that the macromolecular fuse escapes the steric bulk created by the protein globule, collects activation signal in solution, and relays it to the active site of the enzyme. This afforded first-in-class chemical zymogens that are activated via protein-protein interactions. We also document zymogen exchange reactions whereby the polydisulfide is transferred between the interacting proteins via the "chain transfer" bioconjugation mechanism.


Subject(s)
Cysteine , Enzyme Precursors , Cysteine/chemistry , Disulfides/chemistry , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Polymers
6.
Bioconjug Chem ; 33(4): 594-607, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35293739

ABSTRACT

Peptides are prime drug candidates due to their high specificity of action but are disadvantaged by low proteolytic stability. Here, we focus on the development of stabilized analogues of EPI-X4, an endogenous peptide antagonist of CXCR4. We synthesized macromolecular peptide conjugates and performed side-by-side comparison with their albumin-binding counterparts and considered monovalent conjugates, divalent telechelic conjugates, and Y-shaped peptide dimers. All constructs were tested for competition with the CXCR4 antibody-receptor engagement, inhibition of receptor activation, and inhibition of the CXCR4-tropic human immunodeficiency virus infection. We found that the Y-shaped conjugates were more potent than the parent peptide and at the same time more stable in human plasma, with a favorable outlook for translational studies.


Subject(s)
HIV Infections , HIV-1 , Dimerization , HIV-1/physiology , Humans , Peptides/chemistry , Peptides/pharmacology , Receptors, CXCR4/metabolism , Signal Transduction
7.
Acta Biomater ; 101: 422-435, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31669698

ABSTRACT

The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane in order to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. We developed four synthetic approaches to generate RBC-Staphylococcal protein A (RBC-SpA) complexes: amino group targeting through N-hydrosuccinidyl ester-functionalized homobifunctional poly(ethylene glycol) (NHS-PEG-NHS), direct thiol group targeting using heterobifunctional NHS-PEG-maleimide (NHS-PEG-MAL), converted thiol targeting using heterobifunctional NHS-PEG-MAL, and click chemistry using heterobifunctional NHS-PEG-azido (NHS-PEG-N3) and NHS-PEG-alkyne (NHS-PEG-alk). The RBC-PEG-SpA complexes were formed within minutes, followed by the attachment of over 105 antibodies per RBC to the accessible RBC-bound SpA via Fc-Protein A coupling. The RBC-PEG-SpA-antibody arrays were shown to be stable for more than 60 days in PBS and for more than 42 days in serum containing buffer. RBC-PEG-SpA-antibody complexes were shown to remove TNFα from physiological buffer and had similar mechanical properties to unmodified RBCs. Out of the four approaches, the converted thiol method provided the most controlled chemistry and construct stability. We are now ideally positioned to determine the long-term in vivo efficacy of chemically membrane-engineered RBCs to remove antigens, like TNFα, from serum. STATEMENT OF SIGNIFICANCE: The global and economic success of immunoglobulin-based therapeutics in treating a wide range of diseases has heightened the need to further enhance their efficacy and lifetime while diminishing deleterious side effects. The three most ubiquitous challenges of therapeutic immunoglobulin delivery are their relatively short lifetimes in vivo, the immunologic consequences of soluble antibody-antigen complexes, and the emergence of anti-drug antibodies. We describe the rapid, cell-tolerated chemical engineering of the erythrocyte membrane to display any antibody, our model system being the display of anti-Tumor Necrosis Factor (anti-TNFα), on the surface of long-lived red blood cells (RBCs) while masking the antibody's Fc region. Conversion of RBCs into therapeutic delivery vehicles, we argue, would enhance the circulation life of immunoglobulin-based therapeutics while simultaneously evading deleterious immune response.


Subject(s)
Drug Carriers/chemistry , Erythrocytes/metabolism , Immunoglobulins/therapeutic use , Antibodies/metabolism , Antigens/metabolism , Click Chemistry , Erythrocyte Membrane/metabolism , Humans , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Protein Binding , Staphylococcal Protein A/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Sci Rep ; 9(1): 455, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679600

ABSTRACT

The last decade has seen an exponential expansion of interest in conjugating multiple enzymes of cascades in close proximity to each other, with the overarching goal being to accelerate the overall reaction rate. However, some evidence has emerged that there is no effect of proximity channeling on the reaction velocity of the popular GOx-HRP cascade, particularly in the presence of a competing enzyme (catalase). Herein, we rationalize these experimental results quantitatively. We show that, in general, proximity channeling can enhance reaction velocity in the presence of competing enzymes, but in steady state a significant enhancement can only be achieved for diffusion-limited reactions or at high concentrations of competing enzymes. We provide simple equations to estimate the effect of channeling quantitatively and demonstrate that proximity can have a more pronounced effect under crowding conditions in vivo, particularly that crowding can enhance the overall rates of channeled cascade reactions.


Subject(s)
Algorithms , Catalase/metabolism , Glucose Oxidase/metabolism , Horseradish Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Models, Theoretical , Binding, Competitive , Diffusion , Kinetics , Protein Binding , Substrate Specificity
9.
Biomacromolecules ; 19(10): 4044-4051, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30189145

ABSTRACT

Protein-polymer conjugates are powerful combinations of the biotic and abiotic worlds that impact many industries. Predicting the site and impact of polymer growth from the surface of proteins is only useful if we can use that information to choose which site to modify synthetically. We have explored the combination of a predictive algorithm with a unique stepwise atom-transfer radical polymerization (ATRP) to selectively move the predominant modification sites around a model enzyme. Lysozyme was modified with defined stoichiometric ratios of polymerization initiators and initiation inhibitors to selectively and strategically grow poly(carboxybetaine methacrylate) polymers from different protein sites. Electrospray ionization mass spectrometry was used to examine the uniformity of the lysozyme-initiator and lysozyme-inhibitor complexes prior to polymer growth. Bioactivity of the lysozyme-polymer conjugates was examined as a function of polymer location on the enzyme surface. Step-wise atom-transfer radical polymerization from proteins provides a versatile and modular approach that can be extended to the rational and selective design of other protein-polymer conjugates.


Subject(s)
Betaine/chemistry , Muramidase/chemistry , Muramidase/metabolism , Polymers/chemistry , Polymethacrylic Acids/chemistry , Animals , Chickens , Polymerization
10.
Nat Commun ; 9(1): 845, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29487296

ABSTRACT

Facile automated biomacromolecule synthesis is at the heart of blending synthetic and biologic worlds. Full access to abiotic/biotic synthetic diversity first occurred when chemistry was developed to grow nucleic acids and peptides from reversibly immobilized precursors. Protein-polymer conjugates, however, have always been synthesized in solution in multi-step, multi-day processes that couple innovative chemistry with challenging purification. Here we report the generation of protein-polymer hybrids synthesized by protein-ATRP on reversible immobilization supports (PARIS). We utilized modified agarose beads to covalently and reversibly couple to proteins in amino-specific reactions. We then modified reversibly immobilized proteins with protein-reactive ATRP initiators and, after ATRP, we released and analyzed the protein polymers. The activity and stability of PARIS-synthesized and solution-synthesized conjugates demonstrated that PARIS was an effective, rapid, and simple method to generate protein-polymer conjugates. Automation of PARIS significantly reduced synthesis/purification timelines, thereby opening a path to changing how to generate protein-polymer conjugates.


Subject(s)
Polymers/chemical synthesis , Proteins/chemical synthesis , Peptides/chemical synthesis , Peptides/chemistry , Polymerization , Polymers/chemistry , Proteins/chemistry , Solid-Phase Synthesis Techniques
11.
Polym Chem ; 8(2): 375-387, 2017 Jan 14.
Article in English | MEDLINE | ID: mdl-28596807

ABSTRACT

Aqueous supplemental activator and reducing agent atom transfer radical polymerization (SARA ATRP) using inorganic sulfites was successfully carried out for the first time. Under optimized conditions, a well-controlled poly[oligo(ethylene oxide) methyl ether acrylate] (POEOA) was obtained with <30 ppm of soluble copper catalyst using tris(2-pyridylmethyl)amine (TPMA) ligand in the presence of an excess of halide salts (e.g. NaCl). Inorganic sulfites (e.g. Na2S2O4) were continuously fed into the reaction mixture. The mechanistic studies proved that these salts can activate alkyl halides directly and regenerate the activator complex. The effects of the feeding rate of the SARA agent (inorganic sulfites), ligand and its concentration, halide salt and its concentration, sulfite used, and copper concentration, were systematically studied to afford fast polymerizations rates while maintaining the control over polymerization. The kinetic data showed linear first-order kinetics, linear evolution of molecular weights with conversion, and polymers with narrow molecular weight distributions (D ~1.2) during polymerization even at relatively high monomer conversions (~80%). "One-pot" chain extension and "one-pot" block copolymerization experiments proved the high chain-end functionality. The polymerization could be directly regulated by starting or stopping the continuous feeding of the SARA agent. Under biologically relevant conditions, the aqueous SARA ATRP using inorganic sulfites was used to synthesize a well-defined protein-polymer hybrid by grafting of P(OEOA480) from BSA-O-[iBBr]30.

12.
Methods Enzymol ; 590: 347-380, 2017.
Article in English | MEDLINE | ID: mdl-28411645

ABSTRACT

Atom transfer radical polymerization (ATRP) from the surface of a protein can generate remarkably dense polymer shells that serve as armor and rationally tune protein function. Using straightforward chemistry, it is possible to covalently couple or display multiple small molecule initiators onto a protein surface. The chemistry is fine-tuned to be sequence specific (if one desires a single targeted site) at controlled density. Once the initiator is anchored on the protein surface, ATRP is used to grow polymers on protein surface, in situ. The technique is so powerful that a single-protein polymer conjugate molecule can contain more than 90% polymer coating by weight. If desired, stimuli-responsive polymers can be "grown" from the initiated sites to prepare enzyme conjugates that respond to external triggers such as temperature or pH, while still maintaining enzyme activity and stability. Herein, we focus mainly on the synthesis of chymotrypsin-polymer conjugates. Control of the number of covalently coupled initiator sites by changing the stoichiometric ratio between enzyme and the initiator during the synthesis of protein-initiator complexes allowed fine-tuning of the grafting density. For example, very high grafting density chymotrypsin conjugates were prepared from protein-initiator complexes to grow the temperature-responsive polymers, poly(N-isopropylacrylamide), and poly[N,N'-dimethyl(methacryloyloxyethyl) ammonium propane sulfonate]. Controlled growth of polymers from protein surfaces enables one to predictably manipulate enzyme kinetics and stability without the need for molecular biology-dependent mutagenesis.


Subject(s)
Chymotrypsin/chemistry , Enzymes, Immobilized/chemistry , Acrylamides/chemistry , Acrylic Resins/chemistry , Biocatalysis , Enzyme Stability , Kinetics , Polymerization , Proteolysis
13.
Biomacromolecules ; 18(2): 576-586, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28081602

ABSTRACT

The reduced immunogenicity and increased stability of protein-polymer conjugates has made their use in therapeutic applications particularly attractive. However, the physicochemical interactions between polymer and protein, as well as the effect of this interaction on protein activity and stability, are still not fully understood. In this work, polymer-based protein engineering was used to examine the role of polymer physicochemical properties on the activity and stability of the chymotrypsin-polymer conjugates and their degree of binding to intestinal mucin. Four different chymotrypsin-polymer conjugates, each with the same polymer density, were synthesized using "grafting-from" atom transfer radical polymerization. The influence of polymer charge on chymotrypsin-polymer conjugate mucin binding, bioactivity, and stability in stomach acid was determined. Cationic polymers covalently attached to chymotrypsin showed high mucin binding, while zwitterionic, uncharged, and anionic polymers showed no mucin binding. Cationic polymers also increased chymotrypsin activity from pH 6-8, while zwitterionic polymers had no effect, and uncharged and anionic polymers decreased enzyme activity. Lastly, cationic polymers decreased the tendency of chymotrypsin to structurally unfold at extremely low pH, while uncharged and anionic polymers induced unfolding more quickly. We hypothesized that when polymers are covalently attached to the surface of a protein, the degree to which those polymers interact with the protein surface is the predominant determinant of whether the polymer will stabilize or inactivate the protein. Preferential interactions between the polymer and the protein lead to removal of water from the surface of the protein, and this, we believe, inactivates the enzyme.


Subject(s)
Chymotrypsin/metabolism , Gastric Acid/chemistry , Mucins/metabolism , Polymers/metabolism , Cell Adhesion , Chymotrypsin/chemistry , Humans , Mucins/chemistry , Polymerization , Polymers/chemistry , Protein Binding , Protein Engineering
14.
ACS Biomater Sci Eng ; 3(9): 2086-2097, 2017 Sep 11.
Article in English | MEDLINE | ID: mdl-33440561

ABSTRACT

The growth of polymers from the surface of proteins via controlled radical polymerization depends on the attachment of small molecule initiators to amino acid residues. Our ability to control and harness the power of polymer-based protein engineering is reliant on the accuracy of prediction where and how fast atom transfer radical polymerization (ATRP) initiators will react with a protein surface. We performed a systematic characterization of the reaction between a bromine-functionalized N-hydroxysuccinimide amine-reactive ATRP initiator and the amino groups in lysozyme and chymotrypsin. The tertiary structures of the proteins were used to predict computationally α-amino group and lysine side-chain accessibility and analyze the chemical and structural environment of the amino groups. To predict reactivity from accessibility calculations, a probe radius that resembled the size of the initiator molecule was used. Experimental data showed that the rate of initiator-protein modification at each amine site was related to surface accessibility but not the pKa of amino groups. Further refinements of the prediction of where the initiator modified the protein and in what sequence were achieved by considering the local environment of each amino group.

15.
Biosens Bioelectron ; 86: 446-453, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27424262

ABSTRACT

Enzymatic biofuel cells (EBFCs) are capable of generating electricity from physiologically present fuels making them promising power sources for the future of implantable devices. The potential application of such systems is limited, however, by inefficient current generation. Polymer-based protein engineering (PBPE) offers a unique method to tailor enzyme function through tunable modification of the enzyme surface with functional polymers. In this study, we report on the modification of glucose oxidase (GOX) with ferrocene-containing redox polymers to increase current generation efficiency in an enzyme-modified anode. Poly(N-(3-dimethyl(ferrocenyl)methylammonium bromide)propyl acrylamide) (pFcAc) was grown from covalently attached, water-soluble initiator molecules on the surface of GOX in a "grafting-from" approach using atom transfer radical polymerization (ATRP). The covalently-coupled ferrocene-containing polymers on the enzyme surface promoted the effective "wiring" of the GOX active site to an external electrode. The resulting GOX-pFcAc conjugates generated over an order of magnitude increase in current generation efficiency and a 4-fold increase in maximum EBFC power density (≈1.7µWcm(-2)) with similar open circuit voltage (0.27V) compared to native GOX when physically adsorbed onto paddle-shaped electrodes made up of electrospun polyacrylonitrile fibers coated with gold nanoparticles and multi-wall carbon nanotubes. The formation of electroactive enzyme-redox polymer conjugates using PBPE represents a powerful new tool for the improvement of mediated enzyme-based bioelectronics without the need for free redox mediators or anode/cathode compartmentalization.


Subject(s)
Aspergillus niger/enzymology , Bioelectric Energy Sources , Ferrous Compounds/chemistry , Glucose Oxidase/chemistry , Polymers/chemistry , Acrylic Resins/chemistry , Bioelectric Energy Sources/microbiology , Electricity , Electrodes , Metallocenes , Oxidation-Reduction , Polymerization
SELECTION OF CITATIONS
SEARCH DETAIL
...