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










Publication year range
1.
J Am Chem Soc ; 144(23): 10622-10639, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35657057

ABSTRACT

Gram-negative bacteria, especially the ones with multidrug resistance, post dire challenges to antibiotic treatments due to the presence of the outer membrane (OM), which blocks the entry of many antibiotics. Current solutions for such permeability issues, namely lipophilic-cationic derivatization of antibiotics and sensitization with membrane-active agents, cannot effectively potentiate the large, globular, and hydrophilic antibiotics such as vancomycin, due to ineffective disruption of the OM. Here, we present our solution for high-degree OM binding of vancomycin via a hybrid "derivatization-for-sensitization" approach, which features a combination of LPS-targeting lipo-cationic modifications on vancomycin and OM disruption activity from a sensitizing adjuvant. 106- to 107-fold potentiation of vancomycin and 20-fold increase of the sensitizer's effectiveness were achieved with a combination of a vancomycin derivative and its sensitizer. Such potentiation is the result of direct membrane lysis through cooperative membrane binding for the sensitizer-antibiotic complex, which strongly promotes the uptake of vancomycin and adds to the extensive antiresistance effectiveness. The potential of such derivatization-for-sensitization approach was also supported by the combination's potent in vivo antimicrobial efficacy in mouse model studies, and the expanded application of such strategy on other antibiotics and sensitizer structures.


Subject(s)
Gram-Negative Bacteria , Vancomycin , Animals , Anti-Bacterial Agents/pharmacology , Mice , Microbial Sensitivity Tests , Vancomycin/pharmacology
2.
ACS Chem Biol ; 17(2): 414-425, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35129954

ABSTRACT

Site-specific modification of proteins has important applications in biological research and drug development. Reactive tags such as azide, alkyne, and tetrazine have been used extensively to achieve the abovementioned goal. However, bulky side-chain "ligation scars" are often left after the labeling and may hinder the biological application of such engineered protein products. Conjugation chemistry via dehydroalanine (Dha) may provide an opportunity for "traceless" ligation because the activated alkene moiety on Dha can then serve as an electrophile to react with radicalophile, thiol/amine nucleophile, and reactive phosphine probe to introduce a minimal linker in the protein post-translational modifications. In this report, we present a mild and highly efficient enzymatic approach to incorporate Dha with phosphothreonine/serine lyases, OspF and SpvC. These lyases originally catalyze an irreversible elimination reaction that converts a doubly phosphorylated substrate with phosphothreonine (pT) or phosphoserine (pS) to dehydrobutyrine (Dhb) or Dha. To generate a simple monophosphorylated tag for these lyases, we conducted a systematic approach to profile the substrate specificity of OspF and SpvC using peptide arrays and self-assembled monolayers for matrix-assisted laser desorption/ionization mass spectrometry. The optimized tag, [F/Y/W]-pT/pS-[F/Y/W] (where [F/Y/W] indicates an aromatic residue), results in a ∼10-fold enhancement of the overall peptide labeling efficiency via Dha chemistry and enables the first demonstration of protein labeling as well as live cell labeling with a minimal ligation linker via enzyme-mediated incorporation of Dha.


Subject(s)
Lyases , Alanine/analogs & derivatives , Alanine/chemistry , Lyases/metabolism , Phosphothreonine/metabolism , Protein Processing, Post-Translational , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
Biomacromolecules ; 22(6): 2363-2372, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33979120

ABSTRACT

This paper describes the synthesis, characterization, and modeling of a series of molecules having four protein domains attached to a central core. The molecules were assembled with the "megamolecule" strategy, wherein enzymes react with their covalent inhibitors that are substituted on a linker. Three linkers were synthesized, where each had four oligo(ethylene glycol)-based arms terminated in a para-nitrophenyl phosphonate group that is a covalent inhibitor for cutinase. This enzyme is a serine hydrolase and reacts efficiently with the phosphonate to give a new ester linkage at the Ser-120 residue in the active site of the enzyme. Negative-stain transmission electron microscopy (TEM) images confirmed the architecture of the four-armed megamolecules. These cutinase tetramers were also characterized by X-ray crystallography, which confirmed the active-site serine-phosphonate linkage by electron-density maps. Molecular dynamics simulations of the tetracutinase megamolecules using three different force field setups were performed and compared with the TEM observations. Using the Amberff99SB-disp + pH7 force field, the two-dimensional projection distances of the megamolecules were found to agree with the measured dimensions from TEM. The study described here, which combines high-resolution characterization with molecular dynamics simulations, will lead to a comprehensive understanding of the molecular structures and dynamics for this new class of molecules.


Subject(s)
Organophosphonates , Catalytic Domain , Crystallography, X-Ray , Molecular Structure , Protein Domains
4.
Bioconjug Chem ; 32(1): 143-152, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33301672

ABSTRACT

This paper presents a method to synthetically tune atomically precise megamolecule nanobody-enzyme conjugates for prodrug cancer therapy. Previous efforts to create heterobifunctional protein conjugates suffered from heterogeneity in domain stoichiometry, which in part led to the failure of antibody-enzyme conjugates in clinical trials. We used the megamolecule approach to synthesize anti-HER2 nanobody-cytosine deaminase conjugates with tunable numbers of nanobody and enzyme domains in a single, covalent molecule. Linking two nanobody domains to one enzyme domain improved avidity to a human cancer cell line by 4-fold but did not increase cytotoxicity significantly due to lowered enzyme activity. In contrast, a megamolecule composed of one nanobody and two enzyme domains resulted in an 8-fold improvement in the catalytic efficiency and increased the cytotoxic effect by over 5-fold in spheroid culture, indicating that the multimeric structure allowed for an increase in local drug activation. Our work demonstrates that the megamolecule strategy can be used to study structure-function relationships of protein conjugate therapeutics with synthetic control of protein domain stoichiometry.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzymes/chemistry , Prodrugs/therapeutic use , Single-Domain Antibodies/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Humans , Prodrugs/administration & dosage , Proof of Concept Study , Structure-Activity Relationship
5.
J Am Chem Soc ; 142(32): 13657-13661, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32706963

ABSTRACT

This communication describes the design, synthesis, and biological activity of a megamolecule mimic of an anti-HER2 antibody. The antibody mimic was prepared by linking two Fabs from the therapeutic antibody trastuzumab, which are fused through the heavy chain variable domain to either cutinase or SnapTag, with a linker terminated in an irreversible inhibitor for each enzyme. This mimic binds HER2 with comparable avidity to trastuzumab, has similar activity in a cell-based assay, and can arrest tumor growth in a mouse xenograft BT474 tumor model. A panel of 16 bivalent anti-HER2 antibodies were prepared wherein each varied in the orientation of the fusion domain on the Fabs. The analogs displayed a range of cytotoxic activity, and surprisingly, the most active mimic binds to cells with a 10-fold lower avidity than the least active variant suggesting that structure plays a large role in their efficacy. This work suggests that the megamolecule approach can be used to prepare antibody mimics having a broad structural diversity.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Drug Design , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, SCID , Molecular Structure , Trastuzumab/chemistry
6.
J Am Chem Soc ; 142(10): 4534-4538, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32105451

ABSTRACT

This paper presents a solid-phase strategy to efficiently assemble multiprotein scaffolds-known as megamolecules-without the need for protecting groups and with precisely defined nanoscale architectures. The megamolecules are assembled through sequential reactions of linkers that present irreversible inhibitors for enzymes and fusion proteins containing the enzyme domains. Here, a fusion protein containing an N-terminal cutinase and a C-terminal SnapTag domain react with an ethyl p-nitrophenyl phosphonate (pNPP) or a chloro-pyrimidine (CP) group, respectively, to give covalent products. By starting with resin beads that are functionalized with benzylguanine, a series of reactions lead to linear, branched, and dendritic structures that are released from the solid support by addition of TEV protease and that have sizes up to approximately 25 nm.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Macromolecular Substances/chemical synthesis , Recombinant Fusion Proteins/chemistry , Armin/chemistry , Protein Domains , Solid-Phase Synthesis Techniques
7.
J Am Chem Soc ; 140(46): 15731-15743, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30375862

ABSTRACT

In this investigation, we report evidence for energy transfer in new protein-based megamolecules with tunable distances between donor and acceptor fluorescent proteins. The megamolecules used in this work are monodisperse oligomers, with molecular weights of ∼100-300 kDa and lengths of ∼5-20 nm, and are precisely defined structures of fusion protein building blocks and covalent cross-linkers. Such structures are promising because the study of energy transfer in protein complexes is usually difficult in this long length regime due to synthetic limitations. We incorporated fluorescent proteins into the megamolecule structure and varied the separation distance between donor and acceptor by changing the length of the cross-linker in dimer conjugates and inserting nonfluorescent spacer proteins to create oligomers. Two-photon absorption measurements demonstrated strong coupling between donor and acceptor dipoles in the megamolecules. For the dimer systems, no effect of the cross-linker length on energy transfer efficiency was observed with the steady-state fluorescence investigation. However, for the same dimer conjugates, energy transfer rates decreased upon increasing cross-linker length, as evaluated by fluorescence up-conversion. Molecular dynamics simulations were used to rationalize the results, providing quantitative agreement between measured and calculated energy transfer lengths for steady-state results, and showing that the differences between the time-resolved and steady-state measurements arise from the long time scale for large-scale fluctuations in the megamolecule structure. Our results show an increase in energy transfer length with increasing megamolecule size. This is evidence for long-range energy transfer in large protein megamolecules.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Energy Transfer , Fluorescence Resonance Energy Transfer , Molecular Structure
8.
Small ; : e1800923, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29971942

ABSTRACT

This paper describes a microfluidic chip wherein the position and order of two immobilized enzymes affects the type and quantity of reaction products in the flowing fluid. Assembly of the chip is based on a self-assembled monolayer presenting two orthogonal covalent capture ligands that immobilize their respective fusion enzyme. A thiol-tagged substrate is flowed over a region presenting the first enzyme-which generates a product that is efficiently transferred to the second enzyme-and the second enzyme's product binds to an adjacent thiol capture site on the chip. The amount of the three possible reaction products is quantified directly on the chip using self-assembled monolayers for matrix-assisted laser desorption/ionization mass spectrometry, revealing that the same microsystem can be spatiotemporally arranged to produce different products depending on the device design. This work allows for optimizing multistep biochemical transformations in favor of a desired product using a facile reaction and analytical format.

9.
J Am Chem Soc ; 140(20): 6391-6399, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29723476

ABSTRACT

This paper describes the synthesis of giant cyclic molecules having diameters of 10-20 nm. The molecules are prepared through the reactions of a fusion protein building block with small molecule linkers that are terminated in irreversible inhibitors of enzyme domains present in the fusion. This building block has N-terminal cutinase and C-terminal SnapTag domains that react irreversibly with p-nitrophenyl phosphonate (pNPP) and benzylguanine (BG) groups, respectively. We use a bis-BG and a BG-pNPP linker to join these fusion proteins into linear structures that can then react with a bis-pNPP linker that joins the ends into a cyclic product. The last step can occur intramolecularly, to give the macrocycle, or intermolecularly with another equivalent of linker, to give a linear product. Because these are coupled first- and second-order processes, an analysis of product yields from reactions performed at a range of linker concentrations gives rate constants for cyclization. We determined these to be 9.7 × 10-3 s-1, 2.3 × 10-3 s-1, and 8.1 × 10-4 s-1 for the dimer, tetramer, and hexamer, respectively. This work demonstrates an efficient route to cyclic macromolecules having nanoscale dimensions and provides new scaffolds that can be generated using the megamolecule approach.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Guanine/analogs & derivatives , Macrocyclic Compounds/chemistry , Nitrophenols/chemistry , O(6)-Methylguanine-DNA Methyltransferase/chemistry , Organophosphonates/chemistry , Carboxylic Ester Hydrolases/chemical synthesis , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/chemistry , Cyclization , Guanine/chemical synthesis , Macrocyclic Compounds/chemical synthesis , Models, Molecular , Nitrophenols/chemical synthesis , O(6)-Methylguanine-DNA Methyltransferase/chemical synthesis , Organophosphonates/chemical synthesis , Protein Domains , Protein Multimerization
10.
J Am Chem Soc ; 140(17): 5678-5681, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29641892

ABSTRACT

Diabetes affects millions of people worldwide and the number of diagnoses continues to climb annually. Though several effective medications and therapeutic methods have been developed to treat type 1 (T1DM) and type 2 (T2DM) diabetes mellitus, direct insulin injection remains the only effective treatment for insulin resistant (IR) diabetes patients. Here, we immobilize insulin in a crystalline mesoporous metal-organic framework (MOF), NU-1000, and obtain a high loading of ∼40 wt % in only 30 min. The acid-stable MOF capsules are found to effectively prevent insulin from degrading in the presence of stomach acid and the digestive enzyme, pepsin. Furthermore, the encapsulated insulin can be released from NU-1000 under simulated physiological conditions.

12.
Chembiochem ; 13(16): 2331-4, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23070998

ABSTRACT

Enzyme-promoted assembly: The construction of a hetero-bifunctional protein building block, HaloTag-cutinase, that reacts rapidly and selectively with a small-molecule linker is described. The step-wise combination of these building blocks generates a 300 kDa "megamolecule" with precisely defined domain orientation, connectivity, and composition.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Recombinant Fusion Proteins/chemistry , Carboxylic Ester Hydrolases/metabolism , Models, Molecular , Molecular Structure , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism
13.
Nano Lett ; 11(3): 1098-105, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21280643

ABSTRACT

The versatile optical and biological properties of a localized surface plasmon resonance (LSPR) sensor that responds to protein conformational changes are illustrated. The sensor detects conformational changes in a surface-bound construct of the calcium-sensitive protein calmodulin. Increases in calcium concentration induce a 0.96 nm red shift in the spectral position of the LSPR extinction maximum (λ(max)). Addition of a calcium chelating agent forces the protein to return to its original conformation and is detected as a reversal of the λ(max) shift. As opposed to previous work, this work demonstrates that these conformational changes produce a detectable shift in λ(max) even in the absence of a protein label, with a signal:noise ratio near 500. In addition, the protein conformational changes reversibly switch both the wavelength and intensity of the resonance peak, representing an example of a bimodal plasmonic component that simultaneously relays two distinct forms of optical information. This highly versatile plasmonic device acts as a biological sensor, enabling the detection of calcium ions with a biologically relevant limit of detection of 23 µM, as well as the detection of calmodulin-specific protein ligands.


Subject(s)
Biosensing Techniques , Surface Plasmon Resonance/instrumentation , Calmodulin/chemistry , Chelating Agents/chemistry , Ions , Molecular Conformation
14.
J Am Chem Soc ; 130(18): 5836-7, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18402443

ABSTRACT

A plasmonic switch based on the calcium-induced conformational changes of calmodulin is shown to exhibit reversible wavelength modulations in response to changing calcium concentration. The extinction maximum (lambdamax) of a localized surface plasmon resonance (LSPR) sensor functionalized with a novel calmodulin construct, cutinase-calmodulin-cutinase (CutCaMCut), reversibly shifts by 2-3 nm. A high-resolution (HR) LSPR spectrometer with a wavelength resolution (3sigma) of 1.5 x 10-2 nm was developed to detect these wavelength modulations in real-time, providing information about the dynamics and structure of the protein. The rate of conversion from open (Ca2+-bound) to closed (Ca2+-free) calmodulin is shown to be 4-fold faster than the reverse process, with a closing rate of 0.127 s-1 and opening rate of 0.034 s-1. As far as we are aware, this plasmonic switch marks the first use of LSPR spectroscopy to detect reversible conformational changes in an unlabeled protein.


Subject(s)
Calcium/chemistry , Calmodulin/chemistry , Calcium/metabolism , Calmodulin/metabolism , Carboxylic Ester Hydrolases/chemistry , Metal Nanoparticles/chemistry , Nanotechnology , Protein Conformation , Protein Structure, Tertiary , Silver/chemistry , Surface Plasmon Resonance/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...