101
|
Zhu R, Wang S, Xue Z, Han J, Han S. Senescence-associated sialidase revealed by an activatable fluorescence-on labeling probe. Chem Commun (Camb) 2018; 54:11566-11569. [DOI: 10.1039/c8cc07024e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A fluorescence-quenched substrate of sialidase enables fluorescence-on live cell imaging of sialidases, revealing up-regulation of lysosome-associated sialidase in cell senescence.
Collapse
Affiliation(s)
- Rui Zhu
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Siyu Wang
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Zhongwei Xue
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology
- Innovation Center for Cell Signalling Network
- School of Life Sciences
- Xiamen University
- Xiamen
| | - Shoufa Han
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemical Biology
- College of Chemistry and Chemical Engineering
- The Key Laboratory for Chemical Biology of Fujian Province
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation
| |
Collapse
|
102
|
Yu X, Xu J, Zhou Y, Song Q. A facile synthesis of diverse 5-arylated triazoles via a Cu-catalyzed oxidative interrupted click reaction with arylboronic acids in air. Org Chem Front 2018. [DOI: 10.1039/c8qo00590g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A Cu-catalyzed synthesis of 5-arylsubstituted 1,2,3-triazoles via an oxidative interrupted click reaction with arylboronic acids in air at room temperature is disclosed.
Collapse
Affiliation(s)
- Xiaoxia Yu
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- P. R. China
- College of Materials Science & Engineering at Huaqiao University
- Xiamen
| | - Jian Xu
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- P. R. China
| | - Yao Zhou
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- P. R. China
| | - Qiuling Song
- Institute of Next Generation Matter Transformation
- College of Chemical Engineering at Huaqiao University
- P. R. China
| |
Collapse
|
103
|
Cao-Milán R, He LD, Shorkey S, Tonga GY, Wang LS, Zhang X, Uddin I, Das R, Sulak M, Rotello VM. Modulating the Catalytic Activity of Enzyme-like Nanoparticles Through their Surface Functionalization. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2017; 2:624-628. [PMID: 29430303 PMCID: PMC5805145 DOI: 10.1039/c7me00055c] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The inclusion of transition metal catalysts into nanoparticle scaffolds permits the creation of catalytic nanosystems (nanozymes) able to imitate the behaviour of natural enzymes. Here we report the fabrication of a family of nanozymes comprised of bioorthogonal ruthenium catalysts inserted in the protective monolayer of gold nanoparticles. By introducing simple modifications to the functional groups at the surface of the nanozymes, we have demonstrated control over the kinetic mechanism of our system. Cationic nanozymes with hydrophobic surface functionalities tend to replicate the classical Michaelis Menten model, while those with polar groups display substrate inhibition behaviour, a key mechanism present in 20 % of natural enzymes. The structural parameters described herein can be used for creating artificial nanosystems that mimic the complexity observed in cell machinery.
Collapse
Affiliation(s)
- Roberto Cao-Milán
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Luke D He
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Spencer Shorkey
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Gulen Y Tonga
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Li-Sheng Wang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Imad Uddin
- Department of Chemistry, Hazara University, Mansehra 21120, Pakistan
| | - Riddha Das
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| | - Mine Sulak
- School of Applied Science, Pamukkale University, 20600, Çivril, Denizli, Turkey
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| |
Collapse
|
104
|
Zheng YG, Wu XQ, Su J, Jiang P, Xu L, Gao J, Cai B, Ji M. Design and synthesis of a novel photoaffinity probe for labelling EGF receptor tyrosine kinases. J Enzyme Inhib Med Chem 2017; 32:954-959. [PMID: 28718674 PMCID: PMC6009917 DOI: 10.1080/14756366.2017.1344979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) and HER2 are two important tyrosine kinases that play crucial roles in signal transduction pathways that regulate numerous cellular functions including proliferation, differentiation, migration, and angiogenesis. In the past 20 years, many proteomic methods have emerged as powerful methods to evaluate proteins in biological processes and human disease states. Among them, activity-based protein profiling (ABPP) is one useful approach for the functional analysis of proteins. In this study, a novel photoaffinity probe 11 was designed and synthesised to assess the target profiling of the reactive group in the photoaffinity probe 11. Biological evaluation was performed, and the results showed that the novel photoaffinity probe binds to EGFR and HER2 proteins and it hits targets by the reactive group.
Collapse
Affiliation(s)
- You-Guang Zheng
- a College of Pharmacy , Xuzhou Medical University , Xuzhou , PR China
| | - Xiao-Qing Wu
- b Departments of Molecular Biosciences and Radiation Oncology , University of Kansas , Lawrence , KS , USA
| | - Jun Su
- a College of Pharmacy , Xuzhou Medical University , Xuzhou , PR China
| | - Ping Jiang
- a College of Pharmacy , Xuzhou Medical University , Xuzhou , PR China
| | - Liang Xu
- b Departments of Molecular Biosciences and Radiation Oncology , University of Kansas , Lawrence , KS , USA
| | - Jian Gao
- a College of Pharmacy , Xuzhou Medical University , Xuzhou , PR China
| | - Bin Cai
- a College of Pharmacy , Xuzhou Medical University , Xuzhou , PR China
| | - Min Ji
- c School of Chemistry and Chemical Engineering , Southeast University , Nanjing , PR China
| |
Collapse
|
105
|
Ye F, Li J, Yang CG. The development of small-molecule modulators for ClpP protease activity. MOLECULAR BIOSYSTEMS 2017; 13:23-31. [PMID: 27831584 DOI: 10.1039/c6mb00644b] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The global spread of antibiotic resistance among important human pathogens emphasizes the need to find new antibacterial drugs with a novel mode of action. The ClpP protease has been shown to demonstrate its pivotal importance to both the survival and the virulence of pathogenic bacteria during host infection. Deregulating ClpP activity either through overactivation or inhibition could lead to antibacterial activity, declaiming the dual molecular mechanism for small-molecule modulation. Recently, natural products acyldepsipeptides (ADEPs) have been identified as a new class of antibiotics that activate ClpP to a dysfunctional state in the absence of cognate ATPases. ADEPs in combination with rifampicin eradicate deep-seated mouse biofilm infections. In addition, several non-ADEP compounds have been identified as activators of the ClpP proteolytic core without the involvement of ATPases. These findings indicate a general principle for killing dormant cells, the activation and corruption of the ClpP protease, rather than through conventional inhibition. Deletion of the clpP gene reduced the virulence of Staphylococcus aureus, thus making it an ideal antivirulence target. Multiple inhibitors have been developed in order to attenuate the production of extracellular virulence factors of bacteria through covalent modifications on serine in the active site or disruption of oligomerization of ClpP. Interestingly, due to the unusual composition and activation mechanism of ClpP in Mycobacterium tuberculosis, mycobacteria are killed by ADEPs through inhibition of ClpP activity rather than overactivation. In this short review, we will summarize recent progress in the development of small molecules modulating ClpP protease activity for both antibiotics and antivirulence.
Collapse
Affiliation(s)
- Fei Ye
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jiahui Li
- Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Cai-Guang Yang
- Laboratory of Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
106
|
Pan S, Zhang H, Wang C, Yao SCL, Yao SQ. Target identification of natural products and bioactive compounds using affinity-based probes. Nat Prod Rep 2017; 33:612-20. [PMID: 26580476 DOI: 10.1039/c5np00101c] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Covering: 2010 to 2014.Advances in isolation, synthesis and screening strategies have made many bioactive substances available. However, in most cases their putative biological targets remain unknown. Herein, we highlight recent advances in target identification of natural products and bioactive compounds by using affinity-based probes. Aided by photoaffinity labelling, this strategy can capture potential cellular targets (on and off) of a natural product or bioactive compound in live cells directly, even when the compound-target interaction is reversible with moderate affinity. The knowledge of these targets may help uncover molecular pathways and new therapeutics for currently untreatable diseases. In this highlight, we will introduce the development of various photoactivatable groups, their synthesis and applications in target identification of natural products and bioactive compounds, with a focus on work done in recent years and from our laboratory. We will further discuss the strengths and weaknesses of each group and the outlooks for this novel proteome-wide profiling strategy.
Collapse
Affiliation(s)
- Sijun Pan
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| | - Hailong Zhang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| | - Chenyu Wang
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| | - Samantha C L Yao
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore.
| |
Collapse
|
107
|
Zhuang C, Zhang W, Sheng C, Zhang W, Xing C, Miao Z. Chalcone: A Privileged Structure in Medicinal Chemistry. Chem Rev 2017; 117:7762-7810. [PMID: 28488435 PMCID: PMC6131713 DOI: 10.1021/acs.chemrev.7b00020] [Citation(s) in RCA: 847] [Impact Index Per Article: 105.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Privileged structures have been widely used as an effective template in medicinal chemistry for drug discovery. Chalcone is a common simple scaffold found in many naturally occurring compounds. Many chalcone derivatives have also been prepared due to their convenient synthesis. These natural products and synthetic compounds have shown numerous interesting biological activities with clinical potentials against various diseases. This review aims to highlight the recent evidence of chalcone as a privileged scaffold in medicinal chemistry. Multiple aspects of chalcone will be summarized herein, including the isolation of novel chalcone derivatives, the development of new synthetic methodologies, the evaluation of their biological properties, and the exploration of the mechanisms of action as well as target identification. This review is expected to be a comprehensive, authoritative, and critical review of the chalcone template to the chemistry community.
Collapse
Affiliation(s)
- Chunlin Zhuang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Wen Zhang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chunquan Sheng
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Wannian Zhang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
- School of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Drive,
Gainesville, Florida 32610, United States
| | - Zhenyuan Miao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
108
|
Cecioni S, Vocadlo DJ. Carbohydrate Bis-acetal-Based Substrates as Tunable Fluorescence-Quenched Probes for Monitoring exo-Glycosidase Activity. J Am Chem Soc 2017. [PMID: 28631482 DOI: 10.1021/jacs.7b01948] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tunable Förster resonance energy transfer (FRET)-quenched substrates are useful for monitoring the activity of various enzymes within their relevant physiological environments. Development of FRET-quenched substrates for exo-glycosidases, however, has been hindered by their constrained pocket-shaped active sites. Here we report the design of a new class of substrate that overcomes this problem. These Bis-Acetal-Based Substrates (BABS) bear a hemiacetal aglycon leaving group that tethers fluorochromes in close proximity, also positioning them distant from the active site pocket. Following cleavage of the glycosidic bond, the liberated hemiacetal spontaneously breaks down, leading to separation of the fluorophore and quencher. We detail the synthesis and characterization of GlcNAc-BABS, revealing a striking 99.9% quenching efficiency. These substrates are efficiently turned over by the human exo-glycosidase O-GlcNAcase (OGA). We find the hemiacetal leaving group rapidly breaks down, enabling quantitative monitoring of OGA activity. We expect this strategy to be broadly useful for the development of substrate probes for monitoring exo-glycosidases, as well as a range of other enzymes having constrained pocket-shaped active sites.
Collapse
Affiliation(s)
- Samy Cecioni
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia, Canada
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, British Columbia, Canada
| |
Collapse
|
109
|
Ward CC, Kleinman JI, Nomura DK. NHS-Esters As Versatile Reactivity-Based Probes for Mapping Proteome-Wide Ligandable Hotspots. ACS Chem Biol 2017; 12:1478-1483. [PMID: 28445029 DOI: 10.1021/acschembio.7b00125] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Most of the proteome is considered undruggable, oftentimes hindering translational efforts for drug discovery. Identifying previously unknown druggable hotspots in proteins would enable strategies for pharmacologically interrogating these sites with small molecules. Activity-based protein profiling (ABPP) has arisen as a powerful chemoproteomic strategy that uses reactivity-based chemical probes to map reactive, functional, and ligandable hotspots in complex proteomes, which has enabled inhibitor discovery against various therapeutic protein targets. Here, we report an alkyne-functionalized N-hydroxysuccinimide-ester (NHS-ester) as a versatile reactivity-based probe for mapping the reactivity of a wide range of nucleophilic ligandable hotspots, including lysines, serines, threonines, and tyrosines, encompassing active sites, allosteric sites, post-translational modification sites, protein interaction sites, and previously uncharacterized potential binding sites. Surprisingly, we also show that fragment-based NHS-ester ligands can be made to confer selectivity for specific lysine hotspots on specific targets including Dpyd, Aldh2, and Gstt1. We thus put forth NHS-esters as promising reactivity-based probes and chemical scaffolds for covalent ligand discovery.
Collapse
Affiliation(s)
- Carl C. Ward
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Jordan I. Kleinman
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Daniel K. Nomura
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
110
|
Ducharme J, Auclair K. Use of bioconjugation with cytochrome P450 enzymes. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017. [PMID: 28625736 DOI: 10.1016/j.bbapap.2017.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bioconjugation, defined as chemical modification of biomolecules, is widely employed in biological and biophysical studies. It can expand functional diversity and enable applications ranging from biocatalysis, biosensing and even therapy. This review summarizes how chemical modifications of cytochrome P450 enzymes (P450s or CYPs) have contributed to improving our understanding of these enzymes. Genetic modifications of P450s have also proven very useful but are not covered in this review. Bioconjugation has served to gain structural information and investigate the mechanism of P450s via photoaffinity labeling, mechanism-based inhibition (MBI) and fluorescence studies. P450 surface acetylation and protein cross-linking have contributed to the investigation of protein complexes formation involving P450 and its redox partner or other P450 enzymes. Finally, covalent immobilization on polymer surfaces or electrodes has benefited the areas of biocatalysis and biosensor design. This article is part of a Special Issue entitled: Cytochrome P450 biodiversity and biotechnology, edited by Erika Plettner, Gianfranco Gilardi, Luet Wong, Vlada Urlacher, Jared Goldstone.
Collapse
Affiliation(s)
- Julie Ducharme
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Karine Auclair
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada.
| |
Collapse
|
111
|
Zhu M, Harshbarger WD, Robles O, Krysiak J, Hull KG, Cho SW, Richardson RD, Yang Y, Garcia A, Spiegelman L, Ramirez B, Wilson CT, Yau JA, Moore JT, Walker CB, Sacchettini JC, Liu WR, Sieber SA, Smith JW, Romo D. A strategy for dual inhibition of the proteasome and fatty acid synthase with belactosin C-orlistat hybrids. Bioorg Med Chem 2017; 25:2901-2916. [PMID: 28236510 PMCID: PMC5522751 DOI: 10.1016/j.bmc.2017.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/13/2017] [Indexed: 11/21/2022]
Abstract
The proteasome, a validated cellular target for cancer, is central for maintaining cellular homeostasis, while fatty acid synthase (FAS), a novel target for numerous cancers, is responsible for palmitic acid biosynthesis. Perturbation of either enzymatic machine results in decreased proliferation and ultimately cellular apoptosis. Based on structural similarities, we hypothesized that hybrid molecules of belactosin C, a known proteasome inhibitor, and orlistat, a known inhibitor of the thioesterase domain of FAS, could inhibit both enzymes. Herein, we describe proof-of-principle studies leading to the design, synthesis and enzymatic activity of several novel, β-lactone-based, dual inhibitors of these two enzymes. Validation of dual enzyme targeting through activity-based proteome profiling with an alkyne probe modeled after the most potent inhibitor, and preliminary serum stability studies of selected derivatives are also described. These results provide proof of concept for dual targeting of the proteasome and fatty acid synthase-thioesterase (FAS-TE) enabling a new approach for the development of drug-candidates with potential to overcome resistance.
Collapse
Affiliation(s)
- Mingzhao Zhu
- Department of Chemistry & Biochemistry and CPRIT Synthesis and Drug-Lead Discovery Laboratory, Baylor University, Waco, TX 76706, USA
| | - Wayne D Harshbarger
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77840, USA
| | - Omar Robles
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Joanna Krysiak
- Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Kenneth G Hull
- Department of Chemistry & Biochemistry and CPRIT Synthesis and Drug-Lead Discovery Laboratory, Baylor University, Waco, TX 76706, USA
| | - Sung Wook Cho
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | | | - Yanyan Yang
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Andres Garcia
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Lindsey Spiegelman
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Bianca Ramirez
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | | | - Ju Anne Yau
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - James T Moore
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Caitlen B Walker
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - James C Sacchettini
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77840, USA
| | - Wenshe R Liu
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Stephan A Sieber
- Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Jeffrey W Smith
- Sanford Burnham Medical Research Institute, La Jolla, CA, USA
| | - Daniel Romo
- Department of Chemistry & Biochemistry and CPRIT Synthesis and Drug-Lead Discovery Laboratory, Baylor University, Waco, TX 76706, USA.
| |
Collapse
|
112
|
Reactive group-embedded affinity labeling reagent for efficient intracellular protein labeling. Bioorg Med Chem 2017; 25:2888-2894. [DOI: 10.1016/j.bmc.2017.02.059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/26/2017] [Indexed: 01/05/2023]
|
113
|
Kornahrens AF, Cognetta AB, Brody DM, Matthews ML, Cravatt BF, Boger DL. Design of Benzoxathiazin-3-one 1,1-Dioxides as a New Class of Irreversible Serine Hydrolase Inhibitors: Discovery of a Uniquely Selective PNPLA4 Inhibitor. J Am Chem Soc 2017; 139:7052-7061. [PMID: 28498651 PMCID: PMC5501285 DOI: 10.1021/jacs.7b02985] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The design and examination of 4,1,2-benzoxathiazin-3-one 1,1-dioxides as candidate serine hydrolase inhibitors are disclosed, and represent the synthesis and study of a previously unexplored heterocycle. This new class of activated cyclic carbamates provided selective irreversible inhibition of a small subset of serine hydrolases without release of a leaving group, does not covalently modify active site catalytic cysteine and lysine residues of other enzyme classes, and was found to be amenable to predictable structural modifications that modulate intrinsic reactivity or active site recognition. Even more remarkable and within the small pilot series of candidate inhibitors examined in an initial study, an exquisitely selective inhibitor for a poorly characterized serine hydrolase (PNPLA4, patatin-like phospholipase domain-containing protein 4) involved in adipocyte triglyceride homeostasis was discovered.
Collapse
Affiliation(s)
- Anne F. Kornahrens
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| | - Armand B. Cognetta
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| | - Daniel M. Brody
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| | - Megan L. Matthews
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| | - Benjamin F. Cravatt
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037
| |
Collapse
|
114
|
Activity-based proteomics in bioanalysis: past, present and future. Bioanalysis 2017; 9:671-673. [DOI: 10.4155/bio-2017-4999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
115
|
Wang DL, Xiao C, Fu G, Wang X, Li L. Identification of potential serum biomarkers for breast cancer using a functional proteomics technology. Biomark Res 2017; 5:11. [PMID: 28293426 PMCID: PMC5348793 DOI: 10.1186/s40364-017-0092-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/06/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cancer is a genetic disease; its development and metastasis depend on the function of many proteins. Human serum contains thousands of proteins; it is a window for the homeostasis of individual's health. Many of the proteins found in the human serum could be potential biomarkers for cancer early detection and drug efficacy evaluation. METHODS In this study, a functional proteomics technology was used to systematically monitor metabolic enzyme and protease activities from resolved serum proteins produced by a modified 2-D gel separation and subsequent Protein Elution Plate, a method collectively called PEP. All the experiments were repeated at least twice to ensure the validity of the findings. RESULTS For the first time, significant differences were found between breast cancer patient serum and normal serum in two families of enzymes known to be involved in cancer development and metastasis: metabolic enzymes and proteases. Multiple enzyme species were identified in the serum assayed directly or after enrichment. Both qualitative and quantitative differences in the metabolic enzyme and protease activity were detected between breast cancer patient and control group, providing excellent biomarker candidates for breast cancer diagnosis and drug development. CONCLUSIONS This study identified several potential functional protein biomarkers from breast cancer patient serum. It also demonstrated that the functional proteomics technology, PEP, can be applied to the analysis of any functional proteins in human serum which contains thousands of proteins. The study indicated that the functional domain of the human serum could be unlocked with the PEP technology, pointing to a novel alternative for the development of diagnosis biomarkers for breast cancer and other diseases.
Collapse
Affiliation(s)
- David L. Wang
- Department of Biology, Vanderbilt University, Nashville, TN USA
| | | | - Guofeng Fu
- Array Bridge Inc., 4320 Forest Park Ave, Suite 303, St. Louis, MO 63108 USA
| | - Xing Wang
- Array Bridge Inc., 4320 Forest Park Ave, Suite 303, St. Louis, MO 63108 USA
| | - Liang Li
- Zibo Central Hospital, Zibo, China
| |
Collapse
|
116
|
Krysiak J, Stahl M, Vomacka J, Fetzer C, Lakemeyer M, Fux A, Sieber SA. Quantitative Map of β-Lactone-Induced Virulence Regulation. J Proteome Res 2017; 16:1180-1192. [DOI: 10.1021/acs.jproteome.6b00705] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Joanna Krysiak
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Matthias Stahl
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Jan Vomacka
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Christian Fetzer
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Markus Lakemeyer
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Anja Fux
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| | - Stephan A. Sieber
- Department of Chemistry, Chair
of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, D-85747 Garching, Germany
| |
Collapse
|
117
|
Jackson PA, Widen JC, Harki DA, Brummond KM. Covalent Modifiers: A Chemical Perspective on the Reactivity of α,β-Unsaturated Carbonyls with Thiols via Hetero-Michael Addition Reactions. J Med Chem 2017; 60:839-885. [PMID: 27996267 PMCID: PMC5308545 DOI: 10.1021/acs.jmedchem.6b00788] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although Michael acceptors display a potent and broad spectrum of bioactivity, they have largely been ignored in drug discovery because of their presumed indiscriminate reactivity. As such, a dearth of information exists relevant to the thiol reactivity of natural products and their analogues possessing this moiety. In the midst of recently approved acrylamide-containing drugs, it is clear that a good understanding of the hetero-Michael addition reaction and the relative reactivities of biological thiols with Michael acceptors under physiological conditions is needed for the design and use of these compounds as biological tools and potential therapeutics. This Perspective provides information that will contribute to this understanding, such as kinetics of thiol addition reactions, bioactivities, as well as steric and electronic factors that influence the electrophilicity and reversibility of Michael acceptors. This Perspective is focused on α,β-unsaturated carbonyls given their preponderance in bioactive natural products.
Collapse
Affiliation(s)
- Paul A. Jackson
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - John C. Widen
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Daniel A. Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kay M. Brummond
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
118
|
Abstract
Activity-based protein profiling (ABPP) has become a powerful chemoproteomic technology allowing for the dissection of complex ligand-protein interactions in their native cellular environment. One of the biggest challenges for ABPP is the extension of the proteome coverage. In this chapter a new ABPP strategy dedicated to monoamine oxidases (MAO) is presented. These enzymes are representative examples of flavin-dependent oxidases, playing a crucial role in the regulation of nervous system signaling.
Collapse
Affiliation(s)
- Joanna Krysiak
- Chair of Organic Chemistry II, Technische Universität München, Lichtenbergstraße 4, 85748, Garching, Germany
| | - Rolf Breinbauer
- Institute of Organic Chemistry, Graz University of Technology, Stremayrgasse 9, A-8010, Graz, Austria.
| |
Collapse
|
119
|
Tian Y, Wang S, Shang H, Wang M, Sun G, Xu X, Sun X. The proteomic profiling of calenduloside E targets in HUVEC: design, synthesis and application of biotinylated probe BCEA. RSC Adv 2017. [DOI: 10.1039/c6ra25572h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The proteomic profiling of calenduloside E targets was researched by employing the biotinylated probe BCEA of natural product calenduloside E.
Collapse
Affiliation(s)
- Yu Tian
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Shan Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Hai Shang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Xudong Xu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine
- Key Laboratory of efficacy evaluation of Chinese Medicine against glyeolipid metabolism disorder disease, State Administration of Traditional Chinese Medicine
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine
- Ministry of Education, Institute of Medicinal Plant Development
- Chinese Academy of Medical Sciences
| |
Collapse
|
120
|
Schröder SP, van de Sande JW, Kallemeijn WW, Kuo CL, Artola M, van Rooden EJ, Jiang J, Beenakker TJM, Florea BI, Offen WA, Davies GJ, Minnaard AJ, Aerts JMFG, Codée JDC, van der Marel GA, Overkleeft HS. Towards broad spectrum activity-based glycosidase probes: synthesis and evaluation of deoxygenated cyclophellitol aziridines. Chem Commun (Camb) 2017; 53:12528-12531. [DOI: 10.1039/c7cc07730k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Deoxygenated cyclophellitol aziridines enable activity-based inter-class labeling of glycosidases including LC-MS/MS identification.
Collapse
|
121
|
Abstract
Understanding the molecular mechanisms of bacterial pathogenesis and virulence is of great importance from both an academic and clinical perspective, especially in view of an alarming increase in bacterial resistance to existing antibiotics and antibacterial agents. Use of small molecules to dissect the basis of these dynamic processes is a very attractive approach due to their ability for rapid spatiotemporal control of specific biochemical functions. Activity-based protein profiling (ABPP), employing small molecule probes to interrogate enzyme activities in complex proteomes, has emerged as a powerful tool to study bacterial pathogenesis. In this chapter, we present a set of ABPP methods to identify and analyze enzymes essential for growth, metabolism and virulence of different pathogens including S. aureus and L. monocytogenes using natural product-inspired activity-based probes.
Collapse
|
122
|
Hong JA, Choi NE, La YK, Nam HY, Seo J, Lee J. Development of a smart activity-based probe to detect subcellular activity of asparaginyl endopeptidase in living cells. Org Biomol Chem 2017; 15:8018-8022. [DOI: 10.1039/c7ob01467h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A smart activity-based probe that generates a turn-on fluorescence signal in response to enzyme activity was developed, allowing dynamic imaging of subcellular enzyme activity in living cells.
Collapse
Affiliation(s)
- Jong-Ah Hong
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Na-Eun Choi
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Yeo-Kyoung La
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Ho Yeon Nam
- Department of Chemistry
- Gwangju Institute of Science and Technology
- Gwangju 61005
- Republic of Korea
| | - Jiwon Seo
- Department of Chemistry
- Gwangju Institute of Science and Technology
- Gwangju 61005
- Republic of Korea
| | - Jiyoun Lee
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| |
Collapse
|
123
|
Chen X, Wong YK, Wang J, Zhang J, Lee YM, Shen HM, Lin Q, Hua ZC. Target identification with quantitative activity based protein profiling (ABPP). Proteomics 2016; 17. [PMID: 27723264 DOI: 10.1002/pmic.201600212] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/14/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022]
Abstract
As many small bioactive molecules fulfill their functions through interacting with protein targets, the identification of such targets is crucial in understanding their mechanisms of action (MOA) and side effects. With technological advancements in target identification, it has become possible to accurately and comprehensively study the MOA and side effects of small molecules. While small molecules with therapeutic potential were derived solely from nature in the past, the remodeling and synthesis of such molecules have now been made possible. Presently, while some small molecules have seen successful application as drugs, the majority remain undeveloped, requiring further understanding of their MOA and side effects to fully tap into their potential. Given the typical promiscuity of many small molecules and the complexity of the cellular proteome, a high-flux and high-accuracy method is necessary. While affinity chromatography approaches combined with MS have had successes in target identification, limitations associated with nonspecific results remain. To overcome these complications, quantitative chemical proteomics approaches have been developed including metabolic labeling, chemical labeling, and label-free methods. These new approaches are adopted in conjunction with activity-based protein profiling (ABPP), allowing for a rapid process and accurate results. This review will briefly introduce the principles involved in ABPP, then summarize current advances in quantitative chemical proteomics approaches as well as illustrate with examples how ABPP coupled with quantitative chemical proteomics has been used to detect the targets of drugs and other bioactive small molecules including natural products.
Collapse
Affiliation(s)
- Xiao Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China
| | - Yin Kwan Wong
- Department of Biological Sciences, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jigang Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China.,Department of Biological Sciences, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research & Technology (SMART), Singapore
| | - Jianbin Zhang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, P. R., China
| | - Yew-Mun Lee
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, P. R., China
| |
Collapse
|
124
|
Prothiwa M, Szamosvári D, Glasmacher S, Böttcher T. Chemical probes for competitive profiling of the quorum sensing signal synthase PqsD of Pseudomonas aeruginosa. Beilstein J Org Chem 2016; 12:2784-2792. [PMID: 28144351 PMCID: PMC5238532 DOI: 10.3762/bjoc.12.277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022] Open
Abstract
The human pathogen Pseudomonas aeruginosa uses the pqs quorum sensing system to coordinate the production of its broad spectrum of virulence factors to facilitate colonization and infection of its host. Hereby, the enzyme PqsD is a virulence related quorum sensing signal synthase that catalyzes the central step in the biosynthesis of the Pseudomonas quinolone signals HHQ and PQS. We developed a library of cysteine reactive chemical probes with an alkyne handle for fluorescence tagging and report the selective and highly sensitive in vitro labelling of the active site cysteine of this important enzyme. Interestingly, only one type of probe, with a reactive α-chloroacetamide was capable of covalently reacting with the active site. We demonstrated the potential of our probes in a competitive labelling platform where we screened a library of synthetic HHQ and PQS analogues with heteroatom replacements and found several inhibitors of probe binding that may represent promising scaffolds for the development of customized PqsD inhibitors as well as a chemical toolbox to investigate the activity and active site specificity of the enzyme.
Collapse
Affiliation(s)
- Michaela Prothiwa
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Dávid Szamosvári
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Sandra Glasmacher
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Thomas Böttcher
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
125
|
Dormán G, Nakamura H, Pulsipher A, Prestwich GD. The Life of Pi Star: Exploring the Exciting and Forbidden Worlds of the Benzophenone Photophore. Chem Rev 2016; 116:15284-15398. [PMID: 27983805 DOI: 10.1021/acs.chemrev.6b00342] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The widespread applications of benzophenone (BP) photochemistry in biological chemistry, bioorganic chemistry, and material science have been prominent in both academic and industrial research. BP photophores have unique photochemical properties: upon n-π* excitation at 365 nm, a biradicaloid triplet state is formed reversibly, which can abstract a hydrogen atom from accessible C-H bonds; the radicals subsequently recombine, creating a stable covalent C-C bond. This light-directed covalent attachment process is exploited in many different ways: (i) binding/contact site mapping of ligand (or protein)-protein interactions; (ii) identification of molecular targets and interactome mapping; (iii) proteome profiling; (iv) bioconjugation and site-directed modification of biopolymers; (v) surface grafting and immobilization. BP photochemistry also has many practical advantages, including low reactivity toward water, stability in ambient light, and the convenient excitation at 365 nm. In addition, several BP-containing building blocks and reagents are commercially available. In this review, we explore the "forbidden" (transitions) and excitation-activated world of photoinduced covalent attachment of BP photophores by touring a colorful palette of recent examples. In this exploration, we will see the pros and cons of using BP photophores, and we hope that both novice and expert photolabelers will enjoy and be inspired by the breadth and depth of possibilities.
Collapse
Affiliation(s)
- György Dormán
- Targetex llc , Dunakeszi H-2120, Hungary.,Faculty of Pharmacy, University of Szeged , Szeged H-6720, Hungary
| | - Hiroyuki Nakamura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology , Yokohama 226-8503, Japan
| | - Abigail Pulsipher
- GlycoMira Therapeutics, Inc. , Salt Lake City, Utah 84108, United States.,Division of Head and Neck Surgery, Rhinology - Sinus and Skull Base Surgery, Department of Surgery, University of Utah School of Medicine , Salt Lake City, Utah 84108, United States
| | - Glenn D Prestwich
- Division of Head and Neck Surgery, Rhinology - Sinus and Skull Base Surgery, Department of Surgery, University of Utah School of Medicine , Salt Lake City, Utah 84108, United States
| |
Collapse
|
126
|
Romeo E, Pontis S, Ponzano S, Bonezzi F, Migliore M, Di Martino S, Summa M, Piomelli D. Preparation and In Vivo Use of an Activity-based Probe for N-acylethanolamine Acid Amidase. J Vis Exp 2016. [PMID: 27911411 DOI: 10.3791/54652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Activity-based protein profiling (ABPP) is a method for the identification of an enzyme of interest in a complex proteome through the use of a chemical probe that targets the enzyme's active sites. A reporter tag introduced into the probe allows for the detection of the labeled enzyme by in-gel fluorescence scanning, protein blot, fluorescence microscopy, or liquid chromatography-mass spectrometry. Here, we describe the preparation and use of the compound ARN14686, a click chemistry activity-based probe (CC-ABP) that selectively recognizes the enzyme N-acylethanolamine acid amidase (NAAA). NAAA is a cysteine hydrolase that promotes inflammation by deactivating endogenous peroxisome proliferator-activated receptor (PPAR)-alpha agonists such as palmitoylethanolamide (PEA) and oleoylethanolamide (OEA). NAAA is synthesized as an inactive full-length proenzyme, which is activated by autoproteolysis in the acidic pH of the lysosome. Localization studies have shown that NAAA is predominantly expressed in macrophages and other monocyte-derived cells, as well as in B-lymphocytes. We provide examples of how ARN14686 can be used to detect and quantify active NAAA ex vivo in rodent tissues by protein blot and fluorescence microscopy.
Collapse
Affiliation(s)
- Elisa Romeo
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | - Silvia Pontis
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | - Stefano Ponzano
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | - Fabiola Bonezzi
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | - Marco Migliore
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | | | - Maria Summa
- Drug Discovery and Development, Istituto Italiano di Tecnologia
| | - Daniele Piomelli
- Drug Discovery and Development, Istituto Italiano di Tecnologia; Departments of Anatomy and Neurobiology, Pharmacology, and Biological Chemistry, University of California, Irvine School of Medicine;
| |
Collapse
|
127
|
Array-on-Array Strategy For Activity-Based Enzyme Profiling. Methods Mol Biol 2016. [PMID: 27873204 DOI: 10.1007/978-1-4939-6584-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
We describe a novel array on array strategy intended to enhance the throughput of enzymatic activity screening using microarrays. This strategy consists of spotting a first array with large droplets of enzymes with varying concentrations and subsequently spotting a second array with small droplets of fluorogenic substrate on top of the enzyme array. By varying the array on array spotting patterns of different classes of enzyme (e.g., proteases, phosphatases, kinases) and their corresponding fluorogenic substrates, we have the unprecedented ability for testing enzymes and mixed samples in a multiplexed fashion within a single microarray slide. This new approach enables rapid enzyme characterization building upon a one enzyme on one slide droplet-based screening concept previously established.
Collapse
|
128
|
Ono S, Nakai T, Kuroda H, Miyatake R, Horino Y, Abe H, Umezaki M, Oyama H. Site-selective chemical modification of chymotrypsin using peptidyl derivatives bearing optically active diphenyl 1-amino-2-phenylethylphosphonate: Stereochemical effect of the diphenyl phosphonate moiety. Biopolymers 2016; 106:521-30. [PMID: 26615968 DOI: 10.1002/bip.22790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 01/24/2023]
Abstract
Diphenyl (α-aminoalkyl)phosphonates act as mechanism-based inhibitors against serine proteases by forming a covalent bond with the hydroxy group of the active center Ser residue. Because the covalent bond was found to be broken and replaced by 2-pyridinaldoxime methiodide (2PAM), we employed a peptidyl derivative bearing diphenyl 1-amino-2-phenylethylphosphonate moiety (Phe(p) (OPh)2 ) to target the active site of chymotrypsin and to selectively anchor to Lys175 in the vicinity of the active site. Previously, it was reported that the configuration of the α-carbon of phosphorus in diphenyl (α-aminoalkyl)phosphonates affects the inactivation reaction of serine proteases, i.e., the (R)-enantiomeric diphenyl phosphonate is comparable to l-amino acids and it effectively reacts with serine proteases, whereas the (S)-enantiomeric form does not. In this study, we evaluated the stereochemical effect of the phosphonate moiety on the selective chemical modification. Epimeric dipeptidyl derivatives, Ala-(R or S)-Phe(p) (OPh)2 , were prepared by separation with RP-HPLC. A tripeptidyl (R)-epimer (Ala-Ala-(R)-Phe(p) (OPh)2 ) exhibited a more potent inactivation ability against chymotrypsin than the (S)-epimer. The enzyme inactivated by the (R)-epimer was more effectively reactivated with 2PAM than the enzyme inactivated by the (S)-epimer. Finally, N-succinimidyl (NHS) active ester derivatives, NHS-Suc-Ala-Ala- (R or S)-Phe(p) (OPh)2 , were prepared, and we evaluated their action when modifying Lys175 in chymotrypsin. We demonstrated that the epimeric NHS derivative that possessed the diphenyl phosphonate moiety with the (R)-configuration effectively modified Lys175 in chymotrypsin, whereas that with the (S)-configuration did not. These results demonstrate the utility of peptidyl derivatives that bear an optically active diphenyl phosphonate moiety as affinity labeling probes in protein bioconjugation. © 2015 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 521-530, 2016.
Collapse
Affiliation(s)
- Shin Ono
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Hakusan, 924-0838, Japan
| | - Takahiko Nakai
- Graduate School of Science and Engineering, University of Toyama, Toyama, 930-8555, Japan
| | - Hirofumi Kuroda
- Department of General Education, Ishikawa National College of Technology, Ishikawa, 929-0392, Japan
| | - Ryuta Miyatake
- Graduate School of Science and Engineering, University of Toyama, Toyama, 930-8555, Japan
| | - Yoshikazu Horino
- Graduate School of Science and Engineering, University of Toyama, Toyama, 930-8555, Japan
| | - Hitoshi Abe
- Graduate School of Science and Engineering, University of Toyama, Toyama, 930-8555, Japan
| | - Masahito Umezaki
- Institute of Natural Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hiroshi Oyama
- Faculty of Science and Engineering, Setsunan University, Osaka, 572-8508, Japan
| |
Collapse
|
129
|
Mandl FA, Kirsch VC, Ugur I, Kunold E, Vomacka J, Fetzer C, Schneider S, Richter K, Fuchs TM, Antes I, Sieber SA. Naturstoffbasierte Aminoepoxybenzochinone inhibieren das Wachstum verschiedener Serovare des Gram-negativen KrankheitserregersSalmonelladurch Abschwächen der bakteriellen Stressabwehr. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201607338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Franziska A. Mandl
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Volker C. Kirsch
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Ilke Ugur
- Center for Integrated Protein Science; Department für Biowissenschaften; Technische Universität München; Erlenmeyer-Forum 8 85354 Freising Deutschland
| | - Elena Kunold
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Jan Vomacka
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Christian Fetzer
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Sabine Schneider
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Klaus Richter
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Thilo M. Fuchs
- Lehrstuhl für Mikrobielle Ökologie; Zentralinstitut für Ernährungs- und Lebensmittelforschung; Technische Universität München; Weihenstephaner Berg 3 85354 Freising Deutschland
| | - Iris Antes
- Center for Integrated Protein Science; Department für Biowissenschaften; Technische Universität München; Erlenmeyer-Forum 8 85354 Freising Deutschland
| | - Stephan A. Sieber
- Center for Integrated Protein Science, Department Chemie; Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| |
Collapse
|
130
|
Natural‐Product‐Inspired Aminoepoxybenzoquinones Kill Members of the Gram‐Negative Pathogen
Salmonella
by Attenuating Cellular Stress Response. Angew Chem Int Ed Engl 2016; 55:14852-14857. [DOI: 10.1002/anie.201607338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Indexed: 11/07/2022]
|
131
|
Li L, Zhang Z. Development and Applications of the Copper-Catalyzed Azide-Alkyne Cycloaddition (CuAAC) as a Bioorthogonal Reaction. Molecules 2016; 21:E1393. [PMID: 27783053 PMCID: PMC6273301 DOI: 10.3390/molecules21101393] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/14/2016] [Accepted: 10/15/2016] [Indexed: 11/24/2022] Open
Abstract
The emergence of bioorthogonal reactions has greatly broadened the scope of biomolecule labeling and detecting. Of all the bioorthogonal reactions that have been developed, the copper-catalyzed azide-alkyne cycloaddition (CuAAC) is the most widely applied one, mainly because of its relatively fast kinetics and high efficiency. However, the introduction of copper species to in vivo systems raises the issue of potential toxicity. In order to reduce the copper-induced toxicity and further improve the reaction kinetics and efficiency, different strategies have been adopted, including the development of diverse copper chelating ligands to assist the catalytic cycle and the development of chelating azides as reagents. Up to now, the optimization of CuAAC has facilitated its applications in labeling and identifying either specific biomolecule species or on the omics level. Herein, we mainly discuss the efforts in the development of CuAAC to better fit the bioorthogonal reaction criteria and its bioorthogonal applications both in vivo and in vitro.
Collapse
Affiliation(s)
- Li Li
- School of Life Sciences, Peking University, Beijing 100871, China.
- National Institute of Biological Sciences, Beijing 102206, China.
| | - Zhiyuan Zhang
- National Institute of Biological Sciences, Beijing 102206, China.
| |
Collapse
|
132
|
Ando J, Asanuma M, Dodo K, Yamakoshi H, Kawata S, Fujita K, Sodeoka M. Alkyne-Tag SERS Screening and Identification of Small-Molecule-Binding Sites in Protein. J Am Chem Soc 2016; 138:13901-13910. [DOI: 10.1021/jacs.6b06003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Jun Ando
- AMED-CREST, Japan Agency for Medical Research and Development, Saitama 351-0198, Japan
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Synthetic
Organic Chemistry Laboratory, RIKEN, Saitama 351-0198, Japan
- Department
of Applied Physics, Osaka University, Osaka 565-0871, Japan
| | - Miwako Asanuma
- AMED-CREST, Japan Agency for Medical Research and Development, Saitama 351-0198, Japan
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Synthetic
Organic Chemistry Laboratory, RIKEN, Saitama 351-0198, Japan
| | - Kosuke Dodo
- AMED-CREST, Japan Agency for Medical Research and Development, Saitama 351-0198, Japan
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Synthetic
Organic Chemistry Laboratory, RIKEN, Saitama 351-0198, Japan
| | - Hiroyuki Yamakoshi
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Synthetic
Organic Chemistry Laboratory, RIKEN, Saitama 351-0198, Japan
| | - Satoshi Kawata
- Department
of Applied Physics, Osaka University, Osaka 565-0871, Japan
| | - Katsumasa Fujita
- AMED-CREST, Japan Agency for Medical Research and Development, Saitama 351-0198, Japan
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Department
of Applied Physics, Osaka University, Osaka 565-0871, Japan
| | - Mikiko Sodeoka
- AMED-CREST, Japan Agency for Medical Research and Development, Saitama 351-0198, Japan
- Sodeoka
Live Cell Chemistry Project, ERATO, Japan Science and Technology Agency, Saitama 351-0198, Japan
- Synthetic
Organic Chemistry Laboratory, RIKEN, Saitama 351-0198, Japan
| |
Collapse
|
133
|
Irreversible inhibitors of the 3C protease of Coxsackie virus through templated assembly of protein-binding fragments. Nat Commun 2016; 7:12761. [PMID: 27677239 PMCID: PMC5052702 DOI: 10.1038/ncomms12761] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 07/29/2016] [Indexed: 02/01/2023] Open
Abstract
Small-molecule fragments binding to biomacromolecules can be starting points for the development of drugs, but are often difficult to detect due to low affinities. Here we present a strategy that identifies protein-binding fragments through their potential to induce the target-guided formation of covalently bound, irreversible enzyme inhibitors. A protein-binding nucleophile reacts reversibly with a bis-electrophilic warhead, thereby positioning the second electrophile in close proximity of the active site of a viral protease, resulting in the covalent de-activation of the enzyme. The concept is implemented for Coxsackie virus B3 3C protease, a pharmacological target against enteroviral infections. Using an aldehyde-epoxide as bis-electrophile, active fragment combinations are validated through measuring the protein inactivation rate and by detecting covalent protein modification in mass spectrometry. The structure of one enzyme-inhibitor complex is determined by X-ray crystallography. The presented warhead activation assay provides potent non-peptidic, broad-spectrum inhibitors of enteroviral proteases.
Collapse
|
134
|
A highly selective near-infrared fluorescent probe for carboxylesterase 2 and its bioimaging applications in living cells and animals. Biosens Bioelectron 2016; 83:193-9. [DOI: 10.1016/j.bios.2016.04.075] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/18/2016] [Accepted: 04/22/2016] [Indexed: 12/11/2022]
|
135
|
Martell J, Seo Y, Bak DW, Kingsley SF, Tissenbaum HA, Weerapana E. Global Cysteine-Reactivity Profiling during Impaired Insulin/IGF-1 Signaling in C. elegans Identifies Uncharacterized Mediators of Longevity. Cell Chem Biol 2016; 23:955-66. [PMID: 27499530 DOI: 10.1016/j.chembiol.2016.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
In the nematode Caenorhabditis elegans, inactivating mutations in the insulin/IGF-1 receptor, DAF-2, result in a 2-fold increase in lifespan mediated by DAF-16, a FOXO-family transcription factor. Downstream protein activities that directly regulate longevity during impaired insulin/IGF-1 signaling (IIS) are poorly characterized. Here, we use global cysteine-reactivity profiling to identify protein activity changes during impaired IIS. Upon confirming that cysteine reactivity is a good predictor of functionality in C. elegans, we profiled cysteine-reactivity changes between daf-2 and daf-16;daf-2 mutants, and identified 40 proteins that display a >2-fold change. Subsequent RNAi-mediated knockdown studies revealed that lbp-3 and K02D7.1 knockdown caused significant increases in lifespan and dauer formation. The proteins encoded by these two genes, LBP-3 and K02D7.1, are implicated in intracellular fatty acid transport and purine metabolism, respectively. These studies demonstrate that cysteine-reactivity profiling can be complementary to abundance-based transcriptomic and proteomic studies, serving to identify uncharacterized mediators of C. elegans longevity.
Collapse
Affiliation(s)
- Julianne Martell
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Yonghak Seo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel W Bak
- Department of Chemistry, Boston College, Chestnut Hill, MA 02467, USA
| | - Samuel F Kingsley
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heidi A Tissenbaum
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
136
|
Tomohiro T, Nakabayashi M, Sugita Y, Morimoto S. Kinetic controlled affinity labeling of target enzyme with thioester chemistry. Bioorg Med Chem 2016; 24:3336-41. [PMID: 27298000 DOI: 10.1016/j.bmc.2016.05.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/18/2016] [Accepted: 05/28/2016] [Indexed: 11/16/2022]
Abstract
High specificity has been an important feature in affinity labeling for target profiling. Especially, to label targets via rapidly progressing reactions with consumption of ligand (probe), high specificity of reaction with common functional groups of target protein should be achieved without reactions with similar groups of non-target proteins. Herein, we demonstrate the kinetic controlled affinity labeling of acyl CoA synthetase using a fatty acid analogue containing a phenylthioester linkage. High specificity was attained by accelerating the labeling rate in the binding pocket. This approach could be useful for profiling a series of target enzymes and transporters in signal transduction pathways.
Collapse
Affiliation(s)
- Takenori Tomohiro
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2603 Sugitani, Toyama 930-0194, Japan.
| | - Masahiro Nakabayashi
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2603 Sugitani, Toyama 930-0194, Japan
| | - Yuka Sugita
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2603 Sugitani, Toyama 930-0194, Japan
| | - Shota Morimoto
- Laboratory of Biorecognition Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2603 Sugitani, Toyama 930-0194, Japan
| |
Collapse
|
137
|
Abstract
INTRODUCTION Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by expansion of a GAA·TTC triplet in the first intron of the FXN gene, encoding the essential mitochondrial protein frataxin. Repeat expansion results in transcriptional silencing through an epigenetic mechanism, resulting in significant decreases in frataxin protein in affected individuals. Since the FXN protein coding sequence is unchanged in FRDA, an attractive therapeutic approach for this disease would be to increase transcription of pathogenic alleles with small molecules that target the silencing mechanism. AREAS COVERED We review the evidence that histone postsynthetic modifications and heterochromatin formation are responsible for FXN gene silencing in FRDA, along with efforts to reverse silencing with drugs that target histone modifying enzymes. Chemical and pharmacological properties of histone deacetylase (HDAC) inhibitors, which reverse silencing, together with enzyme target profiles and kinetics of inhibition, are discussed. Two HDAC inhibitors have been studied in human clinical trials and the properties of these compounds are compared and contrasted. Efforts to improve on bioavailability, metabolic stability, and target activity are reviewed. EXPERT OPINION 2-aminobenzamide class I HDAC inhibitors are attractive therapeutic small molecules for FRDA. These molecules increase FXN gene expression in human neuronal cells derived from patient induced pluripotent stem cells, and in two mouse models for the disease, as well as in circulating lymphocytes in patients treated in a phase Ib clinical trial. Medicinal chemistry efforts have identified compounds with improved brain penetration, metabolic stability and efficacy in the human neuronal cell model. A clinical candidate will soon be identified for further human testing.
Collapse
Affiliation(s)
- Elisabetta Soragni
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037 USA
| | - Joel M Gottesfeld
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037 USA
| |
Collapse
|
138
|
Mende M, Bednarek C, Wawryszyn M, Sauter P, Biskup MB, Schepers U, Bräse S. Chemical Synthesis of Glycosaminoglycans. Chem Rev 2016; 116:8193-255. [DOI: 10.1021/acs.chemrev.6b00010] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Marco Mende
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, D-76131 Karlsruhe, Germany
| | - Christin Bednarek
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, D-76131 Karlsruhe, Germany
| | - Mirella Wawryszyn
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, D-76131 Karlsruhe, Germany
| | - Paul Sauter
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, D-76131 Karlsruhe, Germany
| | - Moritz B. Biskup
- Division
2—Informatics, Economics and Society, Karlsruhe Institute of Technology (KIT), Kaiserstraße 12, D-76131 Karlsruhe, Germany
| | - Ute Schepers
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, D-76344 Eggenstein-Leopoldshafen, Germany
| | - Stefan Bräse
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, D-76131 Karlsruhe, Germany
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, D-76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
139
|
Jörg M, Scammells PJ. Guidelines for the Synthesis of Small-Molecule Irreversible Probes Targeting G Protein-Coupled Receptors. ChemMedChem 2016; 11:1488-98. [PMID: 27347648 DOI: 10.1002/cmdc.201600066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 06/07/2016] [Indexed: 02/03/2023]
Abstract
Irreversible probes have been proven to be useful pharmacological tools in the study of structural and functional features in drug receptor pharmacology. They have been demonstrated to be particularly valuable for the isolation and purification of receptors. Furthermore, irreversible probes are helpful tools for the identification and characterization of binding sites, thereby supporting the advancement of rational drug design. In this Minireview, we provide insight into universal strategies and guidelines to successfully synthesize irreversible probes that target G protein-coupled receptors (GPCRs). We provide an overview of commonly used chemoreactive and photoreactive groups, and make a comparison of their properties and potential applications. Furthermore, there is a particular focus on synthetic approaches to introduce these reactive groups based on commercially available reagents.
Collapse
Affiliation(s)
- Manuela Jörg
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052, Victoria, Australia.
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, 3052, Victoria, Australia.
| |
Collapse
|
140
|
Chen W, Dong J, Plate L, Mortenson DE, Brighty GJ, Li S, Liu Y, Galmozzi A, Lee PS, Hulce JJ, Cravatt BF, Saez E, Powers ET, Wilson IA, Sharpless KB, Kelly JW. Arylfluorosulfates Inactivate Intracellular Lipid Binding Protein(s) through Chemoselective SuFEx Reaction with a Binding Site Tyr Residue. J Am Chem Soc 2016; 138:7353-64. [PMID: 27191344 PMCID: PMC4909538 DOI: 10.1021/jacs.6b02960] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Arylfluorosulfates have appeared only rarely in the literature and have not been explored as probes for covalent conjugation to proteins, possibly because they were assumed to possess high reactivity, as with other sulfur(VI) halides. However, we find that arylfluorosulfates become reactive only under certain circumstances, e.g., when fluoride displacement by a nucleophile is facilitated. Herein, we explore the reactivity of structurally simple arylfluorosulfates toward the proteome of human cells. We demonstrate that the protein reactivity of arylfluorosulfates is lower than that of the corresponding aryl sulfonyl fluorides, which are better characterized with regard to proteome reactivity. We discovered that simple hydrophobic arylfluorosulfates selectively react with a few members of the intracellular lipid binding protein (iLBP) family. A central function of iLBPs is to deliver small-molecule ligands to nuclear hormone receptors. Arylfluorosulfate probe 1 reacts with a conserved tyrosine residue in the ligand-binding site of a subset of iLBPs. Arylfluorosulfate probes 3 and 4, featuring a biphenyl core, very selectively and efficiently modify cellular retinoic acid binding protein 2 (CRABP2), both in vitro and in living cells. The X-ray crystal structure of the CRABP2-4 conjugate, when considered together with binding site mutagenesis experiments, provides insight into how CRABP2 might activate arylfluorosulfates toward site-specific reaction. Treatment of breast cancer cells with probe 4 attenuates nuclear hormone receptor activity mediated by retinoic acid, an endogenous client lipid of CRABP2. Our findings demonstrate that arylfluorosulfates can selectively target single iLBPs, making them useful for understanding iLBP function.
Collapse
Affiliation(s)
- Wentao Chen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiajia Dong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lars Plate
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David E. Mortenson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel J. Brighty
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Suhua Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yu Liu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Andrea Galmozzi
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter S. Lee
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jonathan J. Hulce
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Enrique Saez
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Evan T. Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - K. Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jeffery W. Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
141
|
Sun Z, Chen X, Wang G, Li L, Fu G, Kuruc M, Wang X. Identification of functional metabolic biomarkers from lung cancer patient serum using PEP technology. Biomark Res 2016; 4:11. [PMID: 27252855 PMCID: PMC4888258 DOI: 10.1186/s40364-016-0065-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Reprogrammed metabolism is a new hallmark of cancer. In many types of cancer, most of the genes in the glycolytic pathway are overexpressed, reflecting an essential shift of metabolism during cancer development. The reprogrammed metabolism contributes to cancer development in multiple ways, from supplying the elevated energy requirement to creating a microenvironment suitable for tumor growth and suppressing the human immune surveillance system. METHOD In this study, a functional proteomics top-down approach was used to systematically monitor metabolic enzyme activities in resolved serum proteins produced by a modified 2-D gel separation and subsequent Protein Elution Plate, a method collectively called PEP. RESULTS We found that the enrichment of low abundance proteins with a bead based product called AlbuVoid™(,) is important to increase the number of observable features and to increase the level of signal achievable from the assay used. From our methods, significant metabolic enzyme activities were detected in both normal and lung cancer patient sera in many fractions after the elution of the 2-D gel separated proteins to the Protein Elution Plate (PEP). Eighteen fractions with the most dramatic metabolic enzyme activity difference between the normal and lung cancer patient sera were submitted for mass spectrometry protein identification. Proteins from the glycolytic metabolic pathway, such as GAPDH along with other proteins not previously annotated to the glycolytic pathway were identified. Further verification with commercially purified GAPDH showed that the addition of purified GAPDH to the metabolic enzyme assay system employed enhanced the enzyme activity, demonstrating that proteins identified from the PEP technology and mass spectrometry could be further verified with biological assay. CONCLUSION This study identified several potential functional enzyme biomarkers from lung cancer patient serum, it provides an alternative and complementary approach to sequence annotation for the discovery of biomarkers in human diseases.
Collapse
Affiliation(s)
- Zhenyu Sun
- />The Third Hospital Affiliated to Nantong University School of Medicine, Wuxi, China
| | - Xiaofeng Chen
- />Shanghai Huashan Hospital, Fudan University School of Medicine, Shanghai, China
| | - Gan. Wang
- />Institute of Environmental Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201 USA
| | - Liang Li
- />Zibo Central Hospital, Zibo, China
| | | | - Matthew Kuruc
- />Biotech Support Group, LLC, Monmouth Junction, NJ USA
| | | |
Collapse
|
142
|
Target identification of natural and traditional medicines with quantitative chemical proteomics approaches. Pharmacol Ther 2016; 162:10-22. [DOI: 10.1016/j.pharmthera.2016.01.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
143
|
Montgomery DC, Garlick JM, Kulkarni RA, Kennedy S, Allali-Hassani A, Kuo YM, Andrews AJ, Wu H, Vedadi M, Meier JL. Global Profiling of Acetyltransferase Feedback Regulation. J Am Chem Soc 2016; 138:6388-91. [PMID: 27149119 DOI: 10.1021/jacs.6b03036] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lysine acetyltransferases (KATs) are key mediators of cell signaling. Methods capable of providing new insights into their regulation thus constitute an important goal. Here we report an optimized platform for profiling KAT-ligand interactions in complex proteomes using inhibitor-functionalized capture resins. This approach greatly expands the scope of KATs, KAT complexes, and CoA-dependent enzymes accessible to chemoproteomic methods. This enhanced profiling platform is then applied in the most comprehensive analysis to date of KAT inhibition by the feedback metabolite CoA. Our studies reveal that members of the KAT superfamily possess a spectrum of sensitivity to CoA and highlight NAT10 as a novel KAT that may be susceptible to metabolic feedback inhibition. This platform provides a powerful tool to define the potency and selectivity of reversible stimuli, such as small molecules and metabolites, that regulate KAT-dependent signaling.
Collapse
Affiliation(s)
- David C Montgomery
- Chemical Biology Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Julie M Garlick
- Chemical Biology Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Rhushikesh A Kulkarni
- Chemical Biology Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States
| | - Steven Kennedy
- Structural Genomics Consortium, University of Toronto , Toronto, Ontario MG5 1L7, Canada
| | | | - Yin-Ming Kuo
- Fox Chase Cancer Institute , Philadelphia, Pennsylvania 19111, United States
| | - Andrew J Andrews
- Fox Chase Cancer Institute , Philadelphia, Pennsylvania 19111, United States
| | - Hong Wu
- Structural Genomics Consortium, University of Toronto , Toronto, Ontario MG5 1L7, Canada
| | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto , Toronto, Ontario MG5 1L7, Canada
| | - Jordan L Meier
- Chemical Biology Laboratory, National Cancer Institute , Frederick, Maryland 21702, United States.,Department of Pharmacology and Toxicology, University of Toronto , Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
144
|
Zhu B, Zhang H, Pan S, Wang C, Ge J, Lee JS, Yao SQ. In Situ Proteome Profiling and Bioimaging Applications of Small-Molecule Affinity-Based Probes Derived From DOT1L Inhibitors. Chemistry 2016; 22:7824-36. [PMID: 27115831 DOI: 10.1002/chem.201600259] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Indexed: 01/01/2023]
Abstract
DOT1L is the sole protein methyltransferase that methylates histone H3 on lysine 79 (H3K79), and is a promising drug target against cancers. Small-molecule inhibitors of DOT1L such as FED1 are potential anti-cancer agents and useful tools to investigate the biological roles of DOT1L in human diseases. FED1 showed excellent in vitro inhibitory activity against DOT1L, but its cellular effect was relatively poor. In this study, we designed and synthesized photo-reactive and "clickable" affinity-based probes (AfBPs), P1 and P2, which were cell-permeable and structural mimics of FED1. The binding and inhibitory effects of these two probes against DOT1L protein were extensively investigated in vitro and in live mammalian cells (in situ). The cellular uptake and sub-cellular localization properties of the probes were subsequently studied in live-cell imaging experiments, and our results revealed that, whereas both P1 and P2 readily entered mammalian cells, most of them were not able to reach the cell nucleus where functional DOT1L resides. This offers a plausible explanation for the poor cellular activity of FED1. Finally with P1/P2, large-scale cell-based proteome profiling, followed by quantitative LC-MS/MS, was carried out to identify potential cellular off-targets of FED1. Amongst the more than 100 candidate off-targets identified, NOP2 (a putative ribosomal RNA methyltransferase) was further confirmed to be likely a genuine off-target of FED1 by preliminary validation experiments including pull-down/Western blotting (PD/WB) and cellular thermal shift assay (CETSA).
Collapse
Affiliation(s)
- Biwei Zhu
- Department of Chemistry, National University of Singapore, 3 Science drive 3, Singapore, 117543, Singapore
| | - Hailong Zhang
- Department of Chemistry, National University of Singapore, 3 Science drive 3, Singapore, 117543, Singapore
| | - Sijun Pan
- Department of Chemistry, National University of Singapore, 3 Science drive 3, Singapore, 117543, Singapore
| | - Chenyu Wang
- Department of Chemistry, National University of Singapore, 3 Science drive 3, Singapore, 117543, Singapore
| | - Jingyan Ge
- Institute of Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, P.R. China
| | - Jun-Seok Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST) and Department of Biological Chemistry, University of Science & Technology, Republic of Korea
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 3 Science drive 3, Singapore, 117543, Singapore.
| |
Collapse
|
145
|
Tuin AW, Mol MAE, van den Berg RM, Fidder A, van der Marel GA, Overkleeft HS, Noort D. Activity-Based Protein Profiling Reveals Broad Reactivity of the Nerve Agent Sarin. Chem Res Toxicol 2016; 22:683-9. [PMID: 19226147 DOI: 10.1021/tx8004218] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Elucidation of noncholinesterase protein targets of organophosphates, and nerve agents in particular, may reveal additional mechanisms for their high toxicity as well as clues for novel therapeutic approaches toward intoxications with these agents. Within this framework, we here describe the synthesis of the activity-based probe 3, which contains a phosphonofluoridate moiety, a P-Me moiety, and a biotinylated O-alkyl group, and its use in activity-based protein profiling with two relevant biological samples, that is, rhesus monkey liver and cultured human A549 lung cells. In this way, we have unearthed eight serine hydrolases (fatty acid synthase, acylpeptide hydrolase, dipeptidyl peptidase 9, prolyl oligopeptidase, carboxylesterase, long-chain acyl coenzyme A thioesterase, PAF acetylhydrolase 1b, and esterase D/S-formyl glutathione hydrolase) as targets that are modified by the nerve agent sarin. It is also shown that the newly developed probe 3 might find its way into the development of alternative, less laborious purification protocols for human butyrylcholinesterase, a potent bioscavenger currently under clinical investigation as a prophylactic/therapeutic for nerve agent intoxications.
Collapse
Affiliation(s)
- Adriaan W Tuin
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands, and Business Unit Biological and Chemical Protection, TNO Defense, Security and Safety, P.O. Box 45, 2280 AA Rijswijk, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
146
|
Chen D, Fan F, Zhao X, Xu F, Chen P, Wang J, Ban L, Liu Z, Feng X, Zhang Y, Liu BF. Single Cell Chemical Proteomics with Membrane-Permeable Activity-Based Probe for Identification of Functional Proteins in Lysosome of Tumors. Anal Chem 2016; 88:2466-71. [DOI: 10.1021/acs.analchem.5b04645] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Dongjuan Chen
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fengkai Fan
- Hubei
Key Laboratory of Purification and Application of Plant Anti-Cancer
Ingredients, College of Chemistry and Life Science, Hubei University of Education, Wuhan, 430205, China
| | - Xingfu Zhao
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fei Xu
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Peng Chen
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jie Wang
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lin Ban
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhihua Liu
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaojun Feng
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yuhui Zhang
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bi-Feng Liu
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics−Hubei Bioinformatics and Molecular Imaging
Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering,
College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
147
|
Abstract
In less than two decades, activity-based protein profiling (ABPP) has expanded to become the de facto tool for the study of small molecule-protein interactions in a proteomic environment. In this issue, Na et al. (2015) present another ABPP method, which they called reactive probe-based chemical proteomics, to study host-pathogen interaction and subsequently identify the protein PheA as a potential key effector during the pathogen infection process.
Collapse
|
148
|
Kowada T, Maeda H, Kikuchi K. BODIPY-based probes for the fluorescence imaging of biomolecules in living cells. Chem Soc Rev 2016; 44:4953-72. [PMID: 25801415 DOI: 10.1039/c5cs00030k] [Citation(s) in RCA: 902] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fluorescence imaging techniques have been widely used to visualize biological molecules and phenomena. In particular, several studies on the development of small-molecule fluorescent probes have been carried out, because their fluorescence properties can be easily tuned by synthetic chemical modification. For this reason, various fluorescent probes have been developed for targeting biological components, such as proteins, peptides, amino acids, and ions, to the interior and exterior of cells. In this review, we cover advances in the development of 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY)-based fluorescent probes for biological studies over the past decade.
Collapse
Affiliation(s)
- Toshiyuki Kowada
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan.
| | | | | |
Collapse
|
149
|
Tiwari VK, Mishra BB, Mishra KB, Mishra N, Singh AS, Chen X. Cu-Catalyzed Click Reaction in Carbohydrate Chemistry. Chem Rev 2016; 116:3086-240. [PMID: 26796328 DOI: 10.1021/acs.chemrev.5b00408] [Citation(s) in RCA: 556] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cu(I)-catalyzed azide-alkyne 1,3-dipolar cycloaddition (CuAAC), popularly known as the "click reaction", serves as the most potent and highly dependable tool for facile construction of simple to complex architectures at the molecular level. Click-knitted threads of two exclusively different molecular entities have created some really interesting structures for more than 15 years with a broad spectrum of applicability, including in the fascinating fields of synthetic chemistry, medicinal science, biochemistry, pharmacology, material science, and catalysis. The unique properties of the carbohydrate moiety and the advantages of highly chemo- and regioselective click chemistry, such as mild reaction conditions, efficient performance with a wide range of solvents, and compatibility with different functionalities, together produce miraculous neoglycoconjugates and neoglycopolymers with various synthetic, biological, and pharmaceutical applications. In this review we highlight the successful advancement of Cu(I)-catalyzed click chemistry in glycoscience and its applications as well as future scope in different streams of applied sciences.
Collapse
Affiliation(s)
- Vinod K Tiwari
- Department of Chemistry, Centre of Advanced Study, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh-221005, India
| | - Bhuwan B Mishra
- Department of Chemistry, Centre of Advanced Study, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh-221005, India
| | - Kunj B Mishra
- Department of Chemistry, Centre of Advanced Study, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh-221005, India
| | - Nidhi Mishra
- Department of Chemistry, Centre of Advanced Study, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh-221005, India
| | - Anoop S Singh
- Department of Chemistry, Centre of Advanced Study, Institute of Science, Banaras Hindu University , Varanasi, Uttar Pradesh-221005, India
| | - Xi Chen
- Department of Chemistry, One Shields Avenue, University of California-Davis , Davis, California 95616, United States
| |
Collapse
|
150
|
Vomacka J, Korotkov VS, Bauer B, Weinandy F, Kunzmann MH, Krysiak J, Baron O, Böttcher T, Lorenz-Baath K, Sieber SA. An Aromatic Hydroxyamide Attenuates MultiresistantStaphylococcus aureusToxin Expression. Chemistry 2016; 22:1622-30. [DOI: 10.1002/chem.201503981] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Indexed: 02/02/2023]
Affiliation(s)
- Jan Vomacka
- Department of Chemistry; AVIRU, GO-Bio-project of the Federal Ministry of Education and Research, FKZ: 031A131; Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Vadim S. Korotkov
- Department of Chemistry; AVIRU, GO-Bio-project of the Federal Ministry of Education and Research, FKZ: 031A131; Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Bianca Bauer
- Department of Chemistry; AVIRU, GO-Bio-project of the Federal Ministry of Education and Research, FKZ: 031A131; Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Franziska Weinandy
- Department of Chemistry; AVIRU, GO-Bio-project of the Federal Ministry of Education and Research, FKZ: 031A131; Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Martin H. Kunzmann
- Department of Chemistry; Chair of Organic Chemistry II; Center for Integrated Protein Science (CIPSM); Institute of Advanced Studies (IAS); Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Joanna Krysiak
- Department of Chemistry; Chair of Organic Chemistry II; Center for Integrated Protein Science (CIPSM); Institute of Advanced Studies (IAS); Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Oliver Baron
- Department of Chemistry; Center for Integrated Protein Science (CIPSM); Ludwig-Maximilians-Universität München (LMU); Butenandtstrasse 5-13 81377 Munich Germany
| | - Thomas Böttcher
- Department of Chemistry; Universität Konstanz; Universitätsstrasse 10 78457 Konstanz Germany
| | - Katrin Lorenz-Baath
- Department of Chemistry; AVIRU, GO-Bio-project of the Federal Ministry of Education and Research, FKZ: 031A131; Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| | - Stephan A. Sieber
- Department of Chemistry; Chair of Organic Chemistry II; Center for Integrated Protein Science (CIPSM); Institute of Advanced Studies (IAS); Technische Universität München (TUM); Lichtenbergstrasse 4 85747 Garching Germany
| |
Collapse
|