1
|
Mando M, Cunningham CW, Grenning AJ. Teaching an Old Dog New Tricks for Achieving CB 2-Selective Inverse Agonism. ACS CENTRAL SCIENCE 2024; 10:946-948. [PMID: 38799666 PMCID: PMC11117306 DOI: 10.1021/acscentsci.4c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Affiliation(s)
- Morgane Mando
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christopher W. Cunningham
- Department
of Pharmaceutical Sciences, Concordia University
Wisconsin, Mequon, Wisconsin 53097, United States
| | - Alexander J. Grenning
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
2
|
Vogt H, Shinkwin P, Huber ME, Staffen N, Hübner H, Gmeiner P, Schiedel M, Weikert D. Development of a Fluorescent Ligand for the Intracellular Allosteric Binding Site of the Neurotensin Receptor 1. ACS Pharmacol Transl Sci 2024; 7:1533-1545. [PMID: 38751637 PMCID: PMC11092115 DOI: 10.1021/acsptsci.4c00086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
The membrane protein family of G protein-coupled receptors (GPCRs) represents a major class of drug targets. Over the last years, the presence of additional intracellular binding sites besides the canonical orthosteric binding pocket has been demonstrated for an increasing number of GPCRs. Allosteric modulators harnessing these pockets may represent valuable alternatives when targeting the orthosteric pocket is not successful for drug development. Starting from SBI-553, a recently discovered intracellular allosteric modulator for neurotensin receptor subtype 1 (NTSR1), we developed the fluorescent molecular probe 14. Compound 14 binds to NTSR1 with an affinity of 0.68 μM in the presence of the agonist NT(8-13). NanoBRET-based ligand binding assays with 14 were established to derive the affinity and structure-activity relationships for allosteric NTSR1 modulators in a direct and nonisotopic manner, thereby facilitating the search for and optimization of novel allosteric NTSR1 ligands. As a consequence of cooperativity between the ligands binding to the allosteric and orthosteric pocket, compound 14 can also be used to investigate orthosteric NTSR1 agonists and antagonists. Moreover, employing 14 as a probe in a drug library screening, we identified novel chemotypes as binders for the intracellular allosteric SBI-553 binding pocket of NTSR1 with single-digit micromolar affinity. These hits may serve as interesting starting points for the development of novel intracellular allosteric ligands for NTSR1 as a highly interesting yet unexploited drug target in the fields of pain and addiction disorder therapy.
Collapse
Affiliation(s)
- Hannah Vogt
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Patrick Shinkwin
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Max E. Huber
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Nico Staffen
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Harald Hübner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Peter Gmeiner
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU
NeW − Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - Matthias Schiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, 38106 Braunschweig, Germany
| | - Dorothee Weikert
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU
NeW − Research Center New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| |
Collapse
|
3
|
Young R, Huang T, Luo Z, Tan YS, Kaur A, Lau YH. Development of stapled NONO-associated peptides reveals unexpected cell permeability and nuclear localisation. J Pept Sci 2024; 30:e3562. [PMID: 38148630 DOI: 10.1002/psc.3562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/28/2023]
Abstract
The non-POU domain-containing octamer-binding protein (NONO) is a nucleic acid-binding protein with diverse functions that has been identified as a potential cancer target in cell biology studies. Little is known about structural motifs that mediate binding to NONO apart from its ability to form homodimers, as well as heterodimers and oligomers with related homologues. We report a stapling approach to macrocyclise helical peptides derived from the insulin-like growth factor binding protein (IGFBP-3) that NONO interacts with, and also from the dimerisation domain of NONO itself. Using a range of chemistries including Pd-catalysed cross-coupling, cysteine arylation and cysteine alkylation, we successfully improved the helicity and observed modest peptide binding to the NONO dimer, although binding could not be saturated at micromolar concentrations. Unexpectedly, we observed cell permeability and preferential nuclear localisation of various dye-labelled peptides in live confocal microscopy, indicating the potential for developing peptide-based tools to study NONO in a cellular context.
Collapse
Affiliation(s)
- Reginald Young
- School of Chemistry, The University of Sydney, Camperdown, Australia
| | - Tiancheng Huang
- School of Chemistry, The University of Sydney, Camperdown, Australia
| | - Zijie Luo
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Matrix, Singapore
| | - Amandeep Kaur
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Monash University, Melbourne, Australia
| | - Yu Heng Lau
- School of Chemistry, The University of Sydney, Camperdown, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Camperdown, Australia
| |
Collapse
|
4
|
Das B, Gupta S, Mondal A, Kalita KJ, Mallick AI, Gupta P. Tuning the Organelle-Specific Imaging and Photodynamic Therapeutic Efficacy of Theranostic Mono- and Trinuclear Organometallic Iridium(III) Complexes. J Med Chem 2023; 66:15550-15563. [PMID: 37950696 DOI: 10.1021/acs.jmedchem.3c01875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2023]
Abstract
The organelle-specific localization of mononuclear and trinuclear iridium(III) complexes and their photodynamic behavior within the cells are described herein, emphasizing their structure-activity relationship. Both the IrA2 and IrB2 complexes possess a pair of phenyl-benzothiazole derived from the -CHO moieties of mononuclear organometallic iridium(III) complexes IrA1 and IrB1, which chelates IrCp*Cl (Cp* = 1,2,3,4,5-pentamethylcyclopentadiene) to afford trinuclear complexes IrA3 and IrB3. Insights into the photophysical and electrochemical parameters of the complexes were obtained by a time-dependent density functional theory study. The synthesized complexes IrA2, IrA3, IrB2, and IrB3 were found to be nontoxic to human MCF7 breast carcinoma cells. However, the photoexcitation of complexes using LED light could effectively trigger intracellular reactive oxygen species (ROS) generation, leading to cell death. Furthermore, to check the organelle-specific localization of IrA2 and IrB2, we observed that both complexes could selectively localize in the endoplasmic reticulum. In contrast, trinuclear IrA3 and IrB3 accumulate in the nuclei. The photoexcitation of complexes using LED light could effectively trigger intracellular reactive oxygen species (ROS) generation, leading to cell death.
Collapse
Affiliation(s)
- Bishnu Das
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Subhadeep Gupta
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Anushka Mondal
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Kalyan Jyoti Kalita
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Amirul Islam Mallick
- Department of Biological Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| | - Parna Gupta
- Department of Chemical Sciences, IISER Kolkata, Mohanpur, Nadia 741246, West Bengal, India
| |
Collapse
|
5
|
Beerkens BL, Snijders IM, Snoeck J, Liu R, Tool ATJ, Le Dévédec SE, Jespers W, Kuijpers TW, van Westen GJ, Heitman LH, IJzerman AP, van der Es D. Development of an Affinity-Based Probe to Profile Endogenous Human Adenosine A3 Receptor Expression. J Med Chem 2023; 66:11399-11413. [PMID: 37531576 PMCID: PMC10461224 DOI: 10.1021/acs.jmedchem.3c00854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 08/04/2023]
Abstract
The adenosine A3 receptor (A3AR) is a G protein-coupled receptor (GPCR) that exerts immunomodulatory effects in pathophysiological conditions such as inflammation and cancer. Thus far, studies toward the downstream effects of A3AR activation have yielded contradictory results, thereby motivating the need for further investigations. Various chemical and biological tools have been developed for this purpose, ranging from fluorescent ligands to antibodies. Nevertheless, these probes are limited by their reversible mode of binding, relatively large size, and often low specificity. Therefore, in this work, we have developed a clickable and covalent affinity-based probe (AfBP) to target the human A3AR. Herein, we show validation of the synthesized AfBP in radioligand displacement, SDS-PAGE, and confocal microscopy experiments as well as utilization of the AfBP for the detection of endogenous A3AR expression in flow cytometry experiments. Ultimately, this AfBP will aid future studies toward the expression and function of the A3AR in pathologies.
Collapse
Affiliation(s)
- Bert L.
H. Beerkens
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Inge M. Snijders
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Joep Snoeck
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Rongfang Liu
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Anton T. J. Tool
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
| | - Sylvia E. Le Dévédec
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Willem Jespers
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Taco W. Kuijpers
- Department
of Molecular Hematology, Sanquin Research, Plesmalaan 125, 1066 CX Amsterdam, The Netherlands
- Department
of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma
Children’s Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gerard J.P. van Westen
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Laura H. Heitman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
- Oncode
Institute, Einsteinweg
55, 2333 CC Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| | - Daan van der Es
- Division
of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333
CC Leiden, The Netherlands
| |
Collapse
|
6
|
Huber ME, Toy L, Schmidt MF, Weikert D, Schiedel M. Small Molecule Tools to Study Cellular Target Engagement for the Intracellular Allosteric Binding Site of GPCRs. Chemistry 2023; 29:e202202565. [PMID: 36193681 PMCID: PMC10100284 DOI: 10.1002/chem.202202565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Indexed: 11/11/2022]
Abstract
A conserved intracellular allosteric binding site (IABS) has recently been identified at several G protein-coupled receptors (GPCRs). Ligands targeting the IABS, so-called intracellular allosteric antagonists, are highly promising compounds for pharmaceutical intervention and currently evaluated in several clinical trials. Beside co-crystal structures that laid the foundation for the structure-based development of intracellular allosteric GPCR antagonists, small molecule tools that enable an unambiguous identification and characterization of intracellular allosteric GPCR ligands are of utmost importance for drug discovery campaigns in this field. Herein, we discuss recent approaches that leverage cellular target engagement studies for the IABS and thus play a critical role in the evaluation of IABS-targeted ligands as potential therapeutic agents.
Collapse
Affiliation(s)
- Max E Huber
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Lara Toy
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Maximilian F Schmidt
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Dorothee Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| | - Matthias Schiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, 91058, Erlangen, Germany
| |
Collapse
|
7
|
Ryan A, Shade O, Bardhan A, Bartnik A, Deiters A. Quantitative Analysis and Optimization of Site-Specific Protein Bioconjugation in Mammalian Cells. Bioconjug Chem 2022; 33:2361-2369. [PMID: 36459098 DOI: 10.1021/acs.bioconjchem.2c00451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Despite a range of covalent protein modifications, few techniques exist for quantification of protein bioconjugation in cells. Here, we describe a novel method for quantifying in cellulo protein bioconjugation through covalent bond formation with HaloTag. This approach utilizes unnatural amino acid (UAA) mutagenesis to selectively install a small and bioorthogonally reactive handle onto the surface of a protein. We utilized the fast kinetics and high selectivity of inverse electron-demand Diels-Alder cycloadditions to evaluate reactions of tetrazine phenylalanine (TetF) with strained trans-cyclooctene-chloroalkane (sTCO-CA) and trans-cyclooctene lysine (TCOK) with tetrazine-chloroalkane (Tet-CA). Following bioconjugation, the chloroalkane ligand is exposed for labeling by the HaloTag enzyme, allowing for straightforward quantification of bioconjugation via simple western blot analysis. We demonstrate the versatility of this tool for quickly and accurately determining the bioconjugation efficiency of different UAA/chloroalkane pairs and for different sites on different proteins of interest, including EGFP and the estrogen-related receptor ERRα.
Collapse
Affiliation(s)
- Amy Ryan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Olivia Shade
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Anirban Bardhan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Aleksander Bartnik
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
8
|
Willén D, Mastio R, Söderlund Z, Manner S, Westergren-Thorsson G, Tykesson E, Ellervik U. Azide-Functionalized Naphthoxyloside as a Tool for Glycosaminoglycan Investigations. Bioconjug Chem 2021; 32:2507-2515. [PMID: 34784477 PMCID: PMC8678990 DOI: 10.1021/acs.bioconjchem.1c00473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/03/2021] [Indexed: 11/28/2022]
Abstract
We present a xylosylated naphthoxyloside carrying a terminal azide functionality that can be used for conjugation using click chemistry. We show that this naphthoxyloside serves as a substrate for β4GalT7 and induces the formation of soluble glycosaminoglycan (GAG) chains with physiologically relevant lengths and sulfation patterns. Finally, we demonstrate its usefulness by conjugation to the Alexa Fluor 647 and TAMRA fluorophores and coupling to a surface plasmon resonance chip for interaction studies with the hepatocyte growth factor known to interact with the GAG heparan sulfate.
Collapse
Affiliation(s)
- Daniel Willén
- Centre
for Analysis and Synthesis, Centre for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | - Roberto Mastio
- Centre
for Analysis and Synthesis, Centre for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | - Zackarias Söderlund
- Department
of Experimental Medical Science, Lund University, P.O. Box 117, SE-221 00 Lund, Sweden
| | - Sophie Manner
- Centre
for Analysis and Synthesis, Centre for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | | | - Emil Tykesson
- Department
of Experimental Medical Science, Lund University, P.O. Box 117, SE-221 00 Lund, Sweden
| | - Ulf Ellervik
- Centre
for Analysis and Synthesis, Centre for Chemistry and Chemical Engineering, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
- Department
of Experimental Medical Science, Lund University, P.O. Box 117, SE-221 00 Lund, Sweden
| |
Collapse
|
9
|
Zeng Y, Huo Y, Yang H. Immunological assays of hemocytes in the Northern Quahog Mercenaria mercenaria. FISH & SHELLFISH IMMUNOLOGY 2021; 118:261-269. [PMID: 34506884 DOI: 10.1016/j.fsi.2021.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 06/13/2023]
Abstract
The northern quahog Mercenaria mercenaria (commonly named hard clam) is an important aquaculture and fishery species along the Atlantic west coast. Environmental stresses, such as heat shock, fluctuating salinity, and harmful algal blooms are major challenges for clam aquaculture. In response to environmental stresses, hemocytes would change dynamically for defense and immunity. The goal of this study was to characterize basic immunological assays of hemocytes in the northern quahog by use of flow cytometry. The objectives were to: 1) develop a non-lethal method for hemolymph collection and dilution; 2) verify the capability of flow cytometry for hemocyte count and type identification through comparison with microscopic observation; 3) validate hemocyte viability assay based on plasma membrane integrity, and 4) develop hemocyte phagocytosis assay by use of fluorescein labeled microbeads. A non-lethal hemocyte collection method was developed using needle insertion through the ligament. Osmolality measurement of serum was the same as that of culture seawater. The pH measurement of serum (7.2) was significantly different from that of culture seawater (8.4). By microscopic observation, three types of hemocytes were identified with granulocytes, the dominant cell type (70 ± 16%), agranulocyte (14 ± 4%), and blast-like cell (16 ± 4%), and no differences were found from the measurements by flow cytometer on FSC/SSC plot (cell size/granularity). The viability of hemocytes based on plasma membrane integrity was 88 ± 6% ranging from 70 to 97% (n = 60, three populations), and viability protocol was further validated with the pre-set expected viability (p ≥ 0.424). Phagocytosis assay of hemocytes with fluorescence beads showed a mean capacity of 10 ± 5% (n = 60, three populations). Incubation time (up to 6 h) or bead concentrations (2:1 or 5:1 to hemocytes) did not affect the phagocytosis measurement. Overall, this study reported the basic characteristics of hemolymph (serum and hemocytes) of northern quahogs. It is expected that the assay methodologies will be applied to evaluation of hemocyte responses to environmental stresses for clam aquaculture.
Collapse
Affiliation(s)
- Yangqing Zeng
- School of Forest, Fisheries, and Geomatics Sciences, Institute of Food and Agricultural Sciences, University of Florida, 7922 NW 71st Street, Gainesville, FL, 32653, USA
| | - Yuanzi Huo
- School of Forest, Fisheries, and Geomatics Sciences, Institute of Food and Agricultural Sciences, University of Florida, 7922 NW 71st Street, Gainesville, FL, 32653, USA
| | - Huiping Yang
- School of Forest, Fisheries, and Geomatics Sciences, Institute of Food and Agricultural Sciences, University of Florida, 7922 NW 71st Street, Gainesville, FL, 32653, USA.
| |
Collapse
|
10
|
van Leeuwen T, Araman C, Pieper Pournara L, Kampstra ASB, Bakkum T, Marqvorsen MHS, Nascimento CR, Groenewold GJM, van der Wulp W, Camps MGM, Janssen GMC, van Veelen PA, van Westen GJP, Janssen APA, Florea BI, Overkleeft HS, Ossendorp FA, Toes REM, van Kasteren SI. Bioorthogonal protein labelling enables the study of antigen processing of citrullinated and carbamylated auto-antigens. RSC Chem Biol 2021; 2:855-862. [PMID: 34212151 PMCID: PMC8190914 DOI: 10.1039/d1cb00009h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 11/21/2022] Open
Abstract
Proteolysis is fundamental to many biological processes. In the immune system, it underpins the activation of the adaptive immune response: degradation of antigenic material into short peptides and presentation thereof on major histocompatibility complexes, leads to activation of T-cells. This initiates the adaptive immune response against many pathogens. Studying proteolysis is difficult, as the oft-used polypeptide reporters are susceptible to proteolytic sequestration themselves. Here we present a new approach that allows the imaging of antigen proteolysis throughout the processing pathway in an unbiased manner. By incorporating bioorthogonal functionalities into the protein in place of methionines, antigens can be followed during degradation, whilst leaving reactive sidechains open to templated and non-templated post-translational modifications, such as citrullination and carbamylation. Using this approach, we followed and imaged the post-uptake fate of the commonly used antigen ovalbumin, as well as the post-translationally citrullinated and/or carbamylated auto-antigen vinculin in rheumatoid arthritis, revealing differences in antigen processing and presentation.
Collapse
Affiliation(s)
- Tyrza van Leeuwen
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Can Araman
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Linda Pieper Pournara
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Arieke S B Kampstra
- Department of Rheumatology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Thomas Bakkum
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Mikkel H S Marqvorsen
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Clarissa R Nascimento
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - G J Mirjam Groenewold
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Willemijn van der Wulp
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Marcel G M Camps
- Department of Immunology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Gerard J P van Westen
- Computational Drug Discovery, Drug Discovery and Safety, LACDR, Leiden University Leiden The Netherlands
| | - Antonius P A Janssen
- Department of Molecular Physiology, Leiden Institute of Chemistry and the Oncode Institute, Leiden University Leiden The Netherlands
| | - Bogdan I Florea
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Herman S Overkleeft
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Ferry A Ossendorp
- Department of Immunology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Sander I van Kasteren
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| |
Collapse
|
11
|
D'Achille AE, Gonzalez-Rodriguez R, Campbell E, Lee BH, Coffer JL, Naumov AV. Rare-Earth-Doped Cerium Oxide Nanocubes for Biomedical Near-Infrared and Magnetic Resonance Imaging. ACS Biomater Sci Eng 2020; 6:6971-6980. [PMID: 33320629 DOI: 10.1021/acsbiomaterials.0c01193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Near-infrared (NIR) fluorescence provides a new avenue for biomedical fluorescence imaging that allows for the tracking of fluorophore through several centimeters of biological tissue. However, such fluorophores are rare and, due to accumulation-derived toxicity, are often restricted from clinical applications. Deep tissue imaging not only provided by near-infrared fluorophores but also conventionally carried out by magnetic resonance imaging (MRI) or computed tomography (CT) is also hampered by the toxicity of the contrast agents. This work offers a biocompatible imaging solution: cerium oxide (CeO2) nanocubes doped with ytterbium or neodymium, and co-doped with gadolinium, showing simultaneous potential for near-infrared (NIR) fluorescence and magnetic resonance imaging (MRI) applications. A synthetic process described in this work allows for the stable incorporation of ytterbium or neodymium, both possessing emissive transitions in the NIR. As a biocompatible nanomaterial, the CeO2 nanocubes act as an ideal host material for doping, minimizing lanthanide fluorescence self-quenching as well as any potential toxicity associated with the dopants. The uptake of nanocubes by HeLa cells maximized at 12 h was monitored by hyperspectral imaging of the ytterbium or neodymium NIR emission, indicating the capacity of the lanthanide-doped nanocubes for in vitro and a potential for in vivo fluorescence imaging. The co-doped nanocubes demonstrate no significant loss of NIR emission intensity upon co-doping with 2 atomic % gadolinium and exhibit magnetic susceptibilities in the range of known negative contrast agents. However, a small increase to 6 atomic % gadolinium significantly affects the magnetic susceptibility ratio (r2/r1), shifting closer to the positive contrast range and suggesting the potential use of the CeO2 nanocube matrix doped with selected rare-earth ions as a tunable MRI contrast agent with NIR imaging capabilities.
Collapse
Affiliation(s)
- Anne E D'Achille
- Department of Chemistry and Biochemistry, Texas Christian University, TCU Box 298860, Fort Worth, Texas 76129, United States
| | - Roberto Gonzalez-Rodriguez
- Department of Chemistry and Biochemistry, Texas Christian University, TCU Box 298860, Fort Worth, Texas 76129, United States
| | - Elizabeth Campbell
- Department of Physics and Astronomy, Texas Christian University, TCU Box 298840, Fort Worth, Texas 76129, United States
| | - Bong Han Lee
- Department of Physics and Astronomy, Texas Christian University, TCU Box 298840, Fort Worth, Texas 76129, United States
| | - Jeffery L Coffer
- Department of Chemistry and Biochemistry, Texas Christian University, TCU Box 298860, Fort Worth, Texas 76129, United States
| | - Anton V Naumov
- Department of Physics and Astronomy, Texas Christian University, TCU Box 298840, Fort Worth, Texas 76129, United States
| |
Collapse
|
12
|
Chen Y, Gao Y, He Y, Zhang G, Wen H, Wang Y, Wu QP, Cui H. Determining Essential Requirements for Fluorophore Selection in Various Fluorescence Applications Taking Advantage of Diverse Structure-Fluorescence Information of Chromone Derivatives. J Med Chem 2020; 64:1001-1017. [PMID: 33307695 DOI: 10.1021/acs.jmedchem.0c01508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Herein, we report our work exploring the essential requirements for fluorophore selection during the development of various fluorescence applications. We assembled a library of chromone-derived fluorophores with diverse structure-fluorescence properties, which allowed us to choose the fluorophore pairs with similar structures but differing fluorescence properties and compared the performance of the selected fluorophore pairs in three types of commonly used fluorescence applications. We found that the selection standard of a suitable fluorophore is variable depending on the application. (1) In fluorescence imaging, fluorophores with strong and constant fluorescence under various conditions, such as a large pH range, are preferred. Notably, (2) in the detection of bioactive species, fluorophores with relatively lower fluorescence quantum yield favor the detection sensitivity. Furthermore, (3) in enzymatic assays employing fluorescence, the key parameter is the binding affinity between the fluorophore and the enzyme.
Collapse
Affiliation(s)
- Yikun Chen
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, No. 8 Liangxiang Donglu, Beijing 102488, China.,State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Yongxin Gao
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Yujun He
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Gang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Hui Wen
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Yuchen Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| | - Qin-Pei Wu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, No. 8 Liangxiang Donglu, Beijing 102488, China
| | - Huaqing Cui
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Xiannongtan Street, Beijing 100050, China
| |
Collapse
|
13
|
Brkovic N, Zhang L, Peters JN, Kleine-Doepke S, Parak WJ, Zhu D. Quantitative Assessment of Endosomal Escape of Various Endocytosed Polymer-Encapsulated Molecular Cargos upon Photothermal Heating. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003639. [PMID: 33108047 DOI: 10.1002/smll.202003639] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Encapsulated molecular cargos are efficiently endocytosed by cells. For cytosolic delivery, understanding the dynamic process of cargos release from the carrier vehicles used for encapsulation and the lysosomes where the carrier vehicles are trapped (which in general is the bottleneck), followed by diffusion in the cytosol is important for improving drug/gene delivery strategies. A methodology is reported to image this process on a millisecond scale and to quantitatively analyze the data. Polyelectrolyte capsules with embedded gold nanostars to encapsulate 43 fluorescent molecular cargos with diverse properties, ranging from small fluorophores to fluorescently labeled proteins, siRNA, etc., are used. By short laser irradiation intracellular release of the molecular cargos from endocytosed capsules into the cytosol is triggered, and their intracellular spreading is imaged. Most of the released molecular cargos evenly distribute inside the entire cell, while others are enriched in certain cell compartments. The time the different molecular cargos take to distribute within cells, i.e., the spreading time, is used as a quantifier. Quantitative analysis reveals that intracellular spread cannot be described by free diffusion, but is determined by interaction of the molecular cargo with intracellular components.
Collapse
Affiliation(s)
- Nico Brkovic
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| | - Li Zhang
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| | - Jan N Peters
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| | - Stephan Kleine-Doepke
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| | - Wolfgang J Parak
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| | - Dingcheng Zhu
- Center for Hybrid Nanostructures (CHyN) and Fachbereich Physik, Universitat Hamburg, Hamburg, 20146, Germany
| |
Collapse
|
14
|
Kim SY, Podder A, Lee H, Cho YJ, Han EH, Khatun S, Sessler JL, Hong KS, Bhuniya S. Self-assembled amphiphilic fluorescent probe: detecting pH-fluctuations within cancer cells and tumour tissues. Chem Sci 2020; 11:9875-9883. [PMID: 34094247 PMCID: PMC8162098 DOI: 10.1039/d0sc03795h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 08/28/2020] [Indexed: 12/26/2022] Open
Abstract
Abnormal anaerobic metabolism leads to a lowering of the pH of many tumours, both within specific intracellular organelles and in the surrounding extracellular regions. Information relating to pH-fluctuations in cells and tissues could aid in the identification of neoplastic lesions and in understanding the determinants of carcinogenesis. Here we report an amphiphilic fluorescent pH probe (CS-1) that, as a result of its temporal motion, provides pH-related information in cancer cell membranes and selected intracellular organelles without the need for specific tumour targeting. Time-dependent cell imaging studies reveal that CS-1 localizes within the cancer cell-membrane about 20 min post-incubation. This is followed by migration to the lysosomes at 30 min before being taken up in the mitochondria after about 60 min. Probe CS-1 can selectively label cancer cells and 3D cancer spheroids and be readily observed using the green fluorescence channel (λ em = 532 nm). In contrast, CS-1 only labels normal cells marginally, with relatively low Pearson's correlation coefficients being found when co-incubated with standard intracellular organelle probes. Both in vivo and ex vivo experiments provide support for the suggestion that CS-1 acts as a fluorescent label for the periphery of tumours, an effect ascribed to proton-induced aggregation. A much lower response is seen for muscle and liver. Based on the present results, we propose that sensors such as CS-1 may have a role to play in the clinical and pathological detection of tumour tissues or serve as guiding aids for surgery.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Cheongju 28119 Korea
| | - Arup Podder
- Amrita Centre for Industrial Research & Innovation, Amrita Vishwa Vidyapeetham Ettimadai Coimbatore 641-112 India
| | - Hyunseung Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Cheongju 28119 Korea
| | - Youn-Joo Cho
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Cheongju 28119 Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Korea
| | - Eun Hee Han
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Cheongju 28119 Korea
| | - Sabina Khatun
- Amrita Centre for Industrial Research & Innovation, Amrita Vishwa Vidyapeetham Ettimadai Coimbatore 641-112 India
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin Austin Texas 78712-1224 USA
| | - Kwan Soo Hong
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute Cheongju 28119 Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Korea
| | - Sankarprasad Bhuniya
- Amrita Centre for Industrial Research & Innovation, Amrita Vishwa Vidyapeetham Ettimadai Coimbatore 641-112 India
- Centre for Interdisciplinary Science, JIS Institute of Advanced Studies and Research, JIS University Kolkata 700-091 India
| |
Collapse
|
15
|
Yoo DY, Barros SA, Brown GC, Rabot C, Bar-Sagi D, Arora PS. Macropinocytosis as a Key Determinant of Peptidomimetic Uptake in Cancer Cells. J Am Chem Soc 2020; 142:14461-14471. [PMID: 32786217 DOI: 10.1021/jacs.0c02109] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptides and peptidomimetics represent the middle space between small molecules and large proteins-they retain the relatively small size and synthetic accessibility of small molecules while providing high binding specificity for biomolecular partners typically observed with proteins. During the course of our efforts to target intracellular protein-protein interactions in cancer, we observed that the cellular uptake of peptides is critically determined by the cell line-specifically, we noted that peptides show better uptake in cancer cells with enhanced macropinocytic indices. Here, we describe the results of our analysis of cellular penetration by different classes of conformationally stabilized peptides. We tested the uptake of linear peptides, peptide macrocycles, stabilized helices, β-hairpin peptides, and cross-linked helix dimers in 11 different cell lines. Efficient uptake of these conformationally defined constructs directly correlated with the macropinocytic activity of each cell line: high uptake of compounds was observed in cells with mutations in certain signaling pathways. Significantly, the study shows that constrained peptides follow the same uptake mechanism as proteins in macropinocytic cells, but unlike proteins, peptide mimics can be readily designed to resist denaturation and proteolytic degradation. Our findings expand the current understanding of cellular uptake in cancer cells by designed peptidomimetics and suggest that cancer cells with certain mutations are suitable mediums for the study of biological pathways with peptide leads.
Collapse
Affiliation(s)
- Daniel Y Yoo
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Stephanie A Barros
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Gordon C Brown
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Christian Rabot
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016, United States
| | - Paramjit S Arora
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
16
|
Ganz D, Harijan D, Wagenknecht HA. Labelling of DNA and RNA in the cellular environment by means of bioorthogonal cycloaddition chemistry. RSC Chem Biol 2020; 1:86-97. [PMID: 34458750 PMCID: PMC8341813 DOI: 10.1039/d0cb00047g] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022] Open
Abstract
Labelling of nucleic acids as biologically important cellular components is a crucial prerequisite for the visualization and understanding of biological processes. Efficient bioorthogonal chemistry and in particular cycloadditions fullfill the requirements for cellular applications. The broadly applied Cu(i)-catalyzed azide-alkyne cycloaddition (CuAAC), however, is limited to labellings in vitro and in fixed cells due to the cytotoxicity of copper salts. Currently, there are three types of copper-free cycloadditions used for nucleic acid labelling in the cellular environment: (i) the ring-strain promoted azide-alkyne cycloaddition (SPAAC), (ii) the "photoclick" 1,3-dipolar cycloadditions, and (iii) the Diels-Alder reactions with inverse electron demand (iEDDA). We review only those building blocks for chemical synthesis on solid phase of DNA and RNA and for enzymatic DNA and RNA preparation, which were applied for labelling of DNA and RNA in situ or in vivo, i.e. in the cellular environment, in fixed or in living cells, by the use of bioorthogonal cycloaddition chemistry. Additionally, we review the current status of orthogonal dual and triple labelling of DNA and RNA in vitro to demonstrate their potential for future applications in situ or in vivo.
Collapse
Affiliation(s)
- Dorothée Ganz
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT) Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| | - Dennis Harijan
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT) Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| | - Hans-Achim Wagenknecht
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT) Fritz-Haber-Weg 6 76131 Karlsruhe Germany
| |
Collapse
|
17
|
Geissinger SE, Schreiber A, Huber MC, Stühn LG, Schiller SM. Adjustable Bioorthogonal Conjugation Platform for Protein Studies in Live Cells Based on Artificial Compartments. ACS Synth Biol 2020; 9:827-842. [PMID: 32130855 DOI: 10.1021/acssynbio.9b00494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The investigation of complex biological processes in vivo often requires defined multiple bioconjugation and positioning of functional entities on 3D structures. Prominent examples include spatially defined protein complexes in nature, facilitating efficient biocatalysis of multistep reactions. Mimicking natural strategies, synthetic scaffolds should comprise bioorthogonal conjugation reactions and allow for absolute stoichiometric quantification as well as facile scalability through scaffold reproduction. Existing in vivo scaffolding strategies often lack covalent conjugations on geometrically confined scaffolds or precise quantitative characterization. Addressing these shortcomings, we present a bioorthogonal dual conjugation platform based on genetically encoded artificial compartments in vivo, comprising two distinct genetically encoded covalent conjugation reactions and their precise stoichiometric quantification. The SpyTag/SpyCatcher (ST/SC) bioconjugation and the controllable strain-promoted azide-alkyne cycloaddition (SPAAC) were implemented on self-assembled protein membrane-based compartments (PMBCs). The SPAAC reaction yield was quantified to be 23% ± 3% and a ST/SC surface conjugation yield of 82% ± 9% was observed, while verifying the compatibility of both chemical reactions as well as enhanced proteolytic stability. Using tandem mass spectrometry, absolute concentrations of the proteinaceous reactants were calculated to be 0.11 ± 0.05 attomol/cell for PMBC surface-tethered mCherry-ST-His and 0.22 ± 0.09 attomol/cell for PMBC-constituting pAzF-SC-E20F20-His. The established in vivo conjugation platform enables quantifiable protein-protein interaction studies on geometrically defined scaffolds and paves the road to investigate effects of scaffold-tethering on enzyme activity.
Collapse
Affiliation(s)
- Süreyya E. Geissinger
- Zentrum für Biosystemanalyse (ZBSA), University of Freiburg, Habsburgerstraße 49, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Andreas Schreiber
- Zentrum für Biosystemanalyse (ZBSA), University of Freiburg, Habsburgerstraße 49, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Matthias C. Huber
- Zentrum für Biosystemanalyse (ZBSA), University of Freiburg, Habsburgerstraße 49, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Lara G. Stühn
- Zentrum für Biosystemanalyse (ZBSA), University of Freiburg, Habsburgerstraße 49, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Stefan M. Schiller
- Zentrum für Biosystemanalyse (ZBSA), University of Freiburg, Habsburgerstraße 49, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Albertstraße 19, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
- Cluster of Excellence livMatS @ FIT, Freiburg Center for Interactive Materials and Bioinspired Technologies, Georges-Köhler-Allee 105, 79110 Freiburg, Germany
| |
Collapse
|
18
|
Practical Guide for Quantification of In Vivo Degradation Rates for Therapeutic Proteins with Single-Cell Resolution Using Fluorescence Ratio Imaging. Pharmaceutics 2020; 12:pharmaceutics12020132. [PMID: 32033318 PMCID: PMC7076450 DOI: 10.3390/pharmaceutics12020132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 01/20/2020] [Accepted: 02/02/2020] [Indexed: 12/20/2022] Open
Abstract
Many tools for studying the pharmacokinetics of biologics lack single-cell resolution to quantify the heterogeneous tissue distribution and subsequent therapeutic degradation in vivo. This protocol describes a dual-labeling technique using two near-infrared dyes with widely differing residualization rates to efficiently quantify in vivo therapeutic protein distribution and degradation rates at the single cell level (number of proteins/cell) via ex vivo flow cytometry and histology. Examples are shown for four biologics with varying rates of receptor internalization and degradation and a secondary dye pair for use in systems with lower receptor expression. Organ biodistribution, tissue-level confocal microscopy, and cellular-level flow cytometry were used to image the multi-scale distribution of these agents in tumor xenograft mouse models. The single-cell measurements reveal highly heterogeneous delivery, and degradation results show the delay between peak tumor uptake and maximum protein degradation. This approach has broad applicability in tracking the tissue and cellular distribution of protein therapeutics for drug development and dose determination.
Collapse
|
19
|
Saha B, Choudhury N, Seal S, Ruidas B, De P. Aromatic Nitrogen Mustard-Based Autofluorescent Amphiphilic Brush Copolymer as pH-Responsive Drug Delivery Vehicle. Biomacromolecules 2018; 20:546-557. [PMID: 30521313 DOI: 10.1021/acs.biomac.8b01468] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Delivery of clinically approved nonfluorescent drugs is facing challenges because it is difficult to monitor the intracellular drug delivery without incorporating any integrated fluorescence moiety into the drug carrier. The present investigation reports the synthesis of a pH-responsive autofluorescent polymeric nanoscaffold for the administration of nonfluorescent aromatic nitrogen mustard chlorambucil (CBL) drug into the cancer cells. Copolymerization of poly(ethylene glycol) (PEG) appended styrene and CBL conjugated N-substituted maleimide monomers enables the formation of well-defined luminescent alternating copolymer. These amphiphilic brush copolymers self-organized in aqueous medium into 25-68 nm nanoparticles, where the CBL drug is enclosed into the core of the self-assembled nanoparticles. In vitro studies revealed ∼70% drug was retained under physiological conditions at pH 7.4 and 37 °C. At endolysosomal pH 5.0, 90% of the CBL was released by the pH-induced cleavage of the aliphatic ester linkages connecting CBL to the maleimide unit. Although the nascent nanoparticle (without drug conjugation) is nontoxic, the drug conjugated nanoparticle showed higher toxicity and superior cell killing capability in cervical cancer (HeLa) cells rather than in normal cells. Interestingly, the copolymer without any conventional chromophore exhibited photoluminescence under UV light irradiation due to the presence of "through-space" π-π interaction between the C═O group of maleimide unit and the adjacent benzene ring of the styrenic monomer. This property helped us intracellular tracking of CBL conjugated autofluorescent nanocarriers through fluorescence microscope imaging. Finally, the 4-(4-nitrobenzyl)pyridine (NBP) colorimetric assay was executed to examine the ability of CBL-based polymeric nanomaterials toward alkylation of DNA.
Collapse
Affiliation(s)
| | | | | | - Bhuban Ruidas
- Centre for Healthcare Science and Technology , Indian Institute of Engineering Science and Technology , Shibpur - 711103 , West Bengal , India
| | | |
Collapse
|
20
|
Roy I, Bobbala S, Zhou J, Nguyen MT, Nalluri SKM, Wu Y, Ferris DP, Scott EA, Wasielewski MR, Stoddart JF. ExTzBox: A Glowing Cyclophane for Live-Cell Imaging. J Am Chem Soc 2018; 140:7206-7212. [PMID: 29771509 DOI: 10.1021/jacs.8b03066] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The ideal fluorescent probe for live-cell imaging is bright and non-cytotoxic and can be delivered easily into the living cells in an efficient manner. The design of synthetic fluorophores having all three of these properties, however, has proved to be challenging. Here, we introduce a simple, yet effective, strategy based on well-established chemistry for designing a new class of fluorescent probes for live-cell imaging. A box-like hybrid cyclophane, namely ExTzBox·4X (6·4X, X = PF6-, Cl-), has been synthesized by connecting an extended viologen (ExBIPY) and a dipyridyl thiazolothiazole (TzBIPY) unit in an end-to-end fashion with two p-xylylene linkers. Photophysical studies show that 6·4Cl has a quantum yield ΦF = 1.00. Furthermore, unlike its ExBIPY2+ and TzBIPY2+ building units, 6·4Cl is non-cytotoxic to RAW 264.7 macrophages, even with a loading concentration as high as 100 μM, presumably on account of its rigid box-like structure which prevents its intercalation into DNA and may inhibit other interactions with it. After gaining an understanding of the toxicity profile of 6·4Cl, we employed it in live-cell imaging. Confocal microscopy has demonstrated that 64+ is taken up by the RAW 264.7 macrophages, allowing the cells to glow brightly with blue laser excitation, without any hint of photobleaching or disruption of normal cell behavior under the imaging conditions. By contrast, the acyclic reference compound Me2TzBIPY·2Cl (4·2Cl) shows very little fluorescence inside the cells, which is quenched completely under the same imaging conditions. In vitro cell investigations underscore the significance of using highly fluorescent box-like rigid cyclophanes for live-cell imaging.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - J Fraser Stoddart
- Institute of Molecular Design and Synthesis , Tianjin University , 92 Weijin Road , Nankai District , Tianjin 300072 , P. R. China
| |
Collapse
|
21
|
Gorka AP, Yamamoto T, Zhu J, Schnermann MJ. Cyanine Photocages Enable Spatial Control of Inducible Cre-Mediated Recombination. Chembiochem 2018; 19:1239-1243. [PMID: 29473264 DOI: 10.1002/cbic.201800061] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Indexed: 12/14/2022]
Abstract
Optical control over protein expression could provide a means to interrogate a range of biological processes. One approach has employed caged ligands of the estrogen receptor (ER) in combination with broadly used ligand-dependent Cre recombinase proteins. Existing approaches use UV or blue wavelengths, which hinders their application in tissue settings. Additionally, issues of payload diffusion can impede fine spatial control over the recombination process. Here, we detail the chemical optimization of a near-infrared (NIR) light-activated variant of the ER antagonist cyclofen. These studies resulted in modification of both the caging group and payload with lipophilic n-butyl esters. The appendage of esters to the cyanine cage improved cellular uptake and retention. The installation of a 4-piperidyl ester enabled high spatial resolution of the light-initiated Cre-mediated recombination event. These studies described chemical modifications with potential general utility for improving spatial control of intracellular caging strategies. Additionally, these efforts will enable future applications to use these molecules in complex physiological settings.
Collapse
Affiliation(s)
- Alexander P Gorka
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA.,Present Address: Department of Chemistry, University of Connecticut, 55 N. Eagleville Road, Storrs, CT, 06269, USA
| | - Tsuyoshi Yamamoto
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA
| | - Jianjian Zhu
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA
| |
Collapse
|
22
|
Yang X, He L, Xu K, Yang Y, Lin W. The development of an ICT-based formaldehyde-responsive fluorescence turn-on probe with a high signal-to-noise ratio. NEW J CHEM 2018. [DOI: 10.1039/c8nj02467g] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An ICT-based formaldehyde fluorescence turn-on probe (PBD-FA) with a high signal-to-noise ratio was judiciously constructed for bio-applications.
Collapse
Affiliation(s)
- Xueling Yang
- Institute of Fluorescent Probes for Biological Imaging
- School of Chemistry and Chemical Engineering
- School of Materials Science and Engineering
- University of Jinan
- Jinan
| | - Longwei He
- Institute of Fluorescent Probes for Biological Imaging
- School of Chemistry and Chemical Engineering
- School of Materials Science and Engineering
- University of Jinan
- Jinan
| | - Kaixin Xu
- Institute of Fluorescent Probes for Biological Imaging
- School of Chemistry and Chemical Engineering
- School of Materials Science and Engineering
- University of Jinan
- Jinan
| | - Yunzhen Yang
- Institute of Fluorescent Probes for Biological Imaging
- School of Chemistry and Chemical Engineering
- School of Materials Science and Engineering
- University of Jinan
- Jinan
| | - Weiying Lin
- Institute of Fluorescent Probes for Biological Imaging
- School of Chemistry and Chemical Engineering
- School of Materials Science and Engineering
- University of Jinan
- Jinan
| |
Collapse
|
23
|
Schiedel M, Herp D, Hammelmann S, Swyter S, Lehotzky A, Robaa D, Oláh J, Ovádi J, Sippl W, Jung M. Chemically Induced Degradation of Sirtuin 2 (Sirt2) by a Proteolysis Targeting Chimera (PROTAC) Based on Sirtuin Rearranging Ligands (SirReals). J Med Chem 2017; 61:482-491. [DOI: 10.1021/acs.jmedchem.6b01872] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Matthias Schiedel
- Institute
of Pharmaceutical Sciences, University of Freiburg, Albertstraße
25, 79104 Freiburg
im Breisgau, Germany
| | - Daniel Herp
- Institute
of Pharmaceutical Sciences, University of Freiburg, Albertstraße
25, 79104 Freiburg
im Breisgau, Germany
| | - Sören Hammelmann
- Institute
of Pharmaceutical Sciences, University of Freiburg, Albertstraße
25, 79104 Freiburg
im Breisgau, Germany
| | - Sören Swyter
- Institute
of Pharmaceutical Sciences, University of Freiburg, Albertstraße
25, 79104 Freiburg
im Breisgau, Germany
| | - Attila Lehotzky
- Institute
of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H 1117 Budapest, Hungary
| | - Dina Robaa
- Institute
of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Straße
4, 06120 Halle (Saale), Germany
| | - Judit Oláh
- Institute
of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H 1117 Budapest, Hungary
| | - Judit Ovádi
- Institute
of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, H 1117 Budapest, Hungary
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Straße
4, 06120 Halle (Saale), Germany
| | - Manfred Jung
- Institute
of Pharmaceutical Sciences, University of Freiburg, Albertstraße
25, 79104 Freiburg
im Breisgau, Germany
- Freiburg
Institute of Advanced Studies (FRIAS), University of Freiburg, Albertstraße
19, 79104 Freiburg
im Breisgau, Germany
| |
Collapse
|
24
|
A Morphological identification cell cytotoxicity assay using cytoplasm-localized fluorescent probe (CLFP) to distinguish living and dead cells. Biochem Biophys Res Commun 2017; 482:257-263. [DOI: 10.1016/j.bbrc.2016.09.169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/29/2016] [Indexed: 01/10/2023]
|
25
|
Yamaguchi K, Murai T, Tsuchiya Y, Miwa Y, Kutsumizu S, Sasamori T, Tokitoh N. Pyridinium 5-aminothiazoles: specific photophysical properties and vapochromism in halogenated solvents. RSC Adv 2017. [DOI: 10.1039/c7ra01896g] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Pyridinium-5-aminothiazoles exhibited bathochromically shifted absorption and fluorescence spectra, solvatochromism, and a reversible vapochromism specific to halogenated solvents.
Collapse
Affiliation(s)
- Kirara Yamaguchi
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Toshiaki Murai
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Yuki Tsuchiya
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Yohei Miwa
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | - Shoichi Kutsumizu
- Department of Chemistry and Biomolecular Science
- Faculty of Engineering
- Gifu University
- Gifu 501-1193
- Japan
| | | | | |
Collapse
|
26
|
Carrillo-Carrion C, Escudero A, Parak WJ. Optical sensing by integration of analyte-sensitive fluorophore to particles. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
27
|
Izabela R, Jarosław R, Magdalena A, Piotr R, Ivan K. Transportan 10 improves the anticancer activity of cisplatin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2016. [PMID: 26899863 DOI: 10.1007/s00210-016-1219-5/figures/8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
The aim of this paper was to examine whether cell-penetrating peptides (CPPs) such as transportan 10 (TP10) or protein transduction domain (PTD4) may improve the anticancer activity of cisplatin (cPt). The complexes of TP10 or PTD4 with cPt were used in the experiments. They were carried out on two non-cancer (HEK293 (human embryonic kidney) and HEL299 (human embryo lung)) and two cancer (HeLa (human cervical cancer) and OS143B (human osteosarcoma 143B)) cell lines. Both complexes were tested (MTT assay) with respect to their anticancer or cytotoxic actions. TAMRA (fluorescent dye)-stained preparations were visualized in a fluorescence microscope. The long-term effect of TP10 + cPt and its components on non-cancer and cancer cell lines was observed in inverted phase contrast microscopy. In the MTT test (cell viability assay), the complex of TP10 + cPt produced a more potent effect on the cancer cell lines (HeLa, OS143B) in comparison to that observed after separate treatment with TP10 or cPt. At the same time, the action of the complex and its components was rather small on non-cancer cell lines. On the other hand, a complex of another CPP with cPt, i.e., PTD4 + cPt, was without a significant effect on the cancer cell line (OS143B). The images of the fluorescent microscopy showed TAMRA-TP10 or TAMRA-TP10 + cPt in the interior of the HeLa cells. In the case of TAMRA-PTD4 or TAMRA-PTD4 + cPt, only the first compound was found inside the cancer cell line. In contrast, none of the tested compounds gained access to the interior of the non-cancer cells (HEK293, HEL299). Long-term incubation with the TP10 + cPt (estimated by inverted phase contrast microscopy) lead to an enhanced action of the complex on cell viability (decrease in the number of cells and change in their morphology) as compared with that produced by each single agent. With regard to the tested CPPs, only TP10 improved the anticancer activity of cisplatin if both compounds were used in the form of a complex. Additionally, the complex was relatively safe for non-cancer cells. What is more, TP10 also produced an anticancer effect on HeLa and OS143B cell lines.
Collapse
Affiliation(s)
- Rusiecka Izabela
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Rekowski Piotr
- Department of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kocić Ivan
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
28
|
Izabela R, Jarosław R, Magdalena A, Piotr R, Ivan K. Transportan 10 improves the anticancer activity of cisplatin. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:485-97. [PMID: 26899863 PMCID: PMC4823340 DOI: 10.1007/s00210-016-1219-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/08/2016] [Indexed: 11/22/2022]
Abstract
The aim of this paper was to examine whether cell-penetrating peptides (CPPs) such as transportan 10 (TP10) or protein transduction domain (PTD4) may improve the anticancer activity of cisplatin (cPt). The complexes of TP10 or PTD4 with cPt were used in the experiments. They were carried out on two non-cancer (HEK293 (human embryonic kidney) and HEL299 (human embryo lung)) and two cancer (HeLa (human cervical cancer) and OS143B (human osteosarcoma 143B)) cell lines. Both complexes were tested (MTT assay) with respect to their anticancer or cytotoxic actions. TAMRA (fluorescent dye)-stained preparations were visualized in a fluorescence microscope. The long-term effect of TP10 + cPt and its components on non-cancer and cancer cell lines was observed in inverted phase contrast microscopy. In the MTT test (cell viability assay), the complex of TP10 + cPt produced a more potent effect on the cancer cell lines (HeLa, OS143B) in comparison to that observed after separate treatment with TP10 or cPt. At the same time, the action of the complex and its components was rather small on non-cancer cell lines. On the other hand, a complex of another CPP with cPt, i.e., PTD4 + cPt, was without a significant effect on the cancer cell line (OS143B). The images of the fluorescent microscopy showed TAMRA-TP10 or TAMRA-TP10 + cPt in the interior of the HeLa cells. In the case of TAMRA-PTD4 or TAMRA-PTD4 + cPt, only the first compound was found inside the cancer cell line. In contrast, none of the tested compounds gained access to the interior of the non-cancer cells (HEK293, HEL299). Long-term incubation with the TP10 + cPt (estimated by inverted phase contrast microscopy) lead to an enhanced action of the complex on cell viability (decrease in the number of cells and change in their morphology) as compared with that produced by each single agent. With regard to the tested CPPs, only TP10 improved the anticancer activity of cisplatin if both compounds were used in the form of a complex. Additionally, the complex was relatively safe for non-cancer cells. What is more, TP10 also produced an anticancer effect on HeLa and OS143B cell lines.
Collapse
Affiliation(s)
- Rusiecka Izabela
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | - Rekowski Piotr
- Department of Chemistry, University of Gdańsk, Gdańsk, Poland
| | - Kocić Ivan
- Department of Pharmacology, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
29
|
Hou Y, Arai S, Kitaguchi T, Suzuki M. Intracellular bottom-up generation of targeted nanosensors for single-molecule imaging. NANOSCALE 2016; 8:3218-3225. [PMID: 26731153 DOI: 10.1039/c5nr08012f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Organic dyes are useful tools for sensing cellular activities but unfavorable in single-molecule imaging, whereas quantum dots (QDs) are widely applied in single-molecule imaging but with few sensing applications. Here, to visualize cellular activities by monitoring the response of a single probe in living cells, we propose a bottom-up approach to generate nanoprobes where four organic dyes are conjugated to tetravalent single-chain avidin (scAVD) proteins via an intracellular click reaction. We demonstrate that the nanoprobes, exhibiting increased brightness and enhanced photostability, were detectable as single dots in living cells. The ease of intracellular targeting allowed the tracking of endoplasmic reticulum (ER) remodeling with nanometer spatial resolution. Conjugating thermosensitive dyes generated temperature-sensitive nanoprobes on ER membranes that successfully monitored local temperature changes in response to external heat pulses. Our approach is potentially a suitable tool for visualizing localized cellular activities with single probe sensitivity in living cells.
Collapse
Affiliation(s)
- Yanyan Hou
- WASEDA Bioscience Research Institute in Singapore (WABIOS), 11 Biopolis Way, #05-02 Helios, Singapore 138667, Singapore.
| | | | | | | |
Collapse
|
30
|
Meimetis LG, Boros E, Carlson JC, Ran C, Caravan P, Weissleder R. Bioorthogonal Fluorophore Linked DFO-Technology Enabling Facile Chelator Quantification and Multimodal Imaging of Antibodies. Bioconjug Chem 2016; 27:257-63. [PMID: 26684717 PMCID: PMC4858350 DOI: 10.1021/acs.bioconjchem.5b00630] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Herein we describe the development and application of a bioorthogonal fluorogenic chelate linker that can be used for facile creation of labeled imaging agents. The chelate linker is based on the trans-cyclooctene(TCO)-tetrazine(Tz) chemistry platform and incorporates deferoxamine (DFO) as a (89)Zr PET tracer and a BODIPY fluorophore for multimodal imaging. The rapid (<3 min) ligation between mAb-TCO and Tz-BODIPY-DFO chelator is monitored using fluorescence and allows for determination of labeling completion. Utilizing BODIPY as the linker between mAb and DFO facilitates in chelator quantification using spectrophotometry, allowing for an alternative to traditional methods (mass and isotope dilution assay). Radiolabeling with (89)Zr to form (89)Zr-DFO-BODIPY-trastuzumab was found to be quantitative after incubation at room temperature for 1 h (1.5 mCi/mg specific activity). The cell binding assay using HER2+ (BT474) and HER2- (BT20) cell lines showed significant binding to (89)Zr-DFO-BODIPY-trastuzumab (6.45 ± 1.87% in BT474 versus 1.47 ± 0.39% in BT20). In vivo PET imaging of mice bearing BT20 or BT474 xenografts with (89)Zr-DFO-BODIPY-trastuzumab showed high tumor conspicuity, and biodistribution confirmed excellent, specific probe uptake of 237.3 ± 14.5% ID/g in BT474 xenografts compared to low, nonspecific probe uptake in BT20 xenografts (16.4 ± 5.6% ID/g) 96 h p.i. . Ex vivo fluorescence (465ex/520em) of selected tissues confirmed superb target localization and persistence of the fluorescence of (89)Zr-DFO-BODIPY-trastuzumab. The described platform is universally adaptable for simple antibody labeling.
Collapse
Affiliation(s)
- Labros G. Meimetis
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Eszter Boros
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 Thirteenth street, Charlestown, Massachusetts 02129, United States
| | - Jonathan C. Carlson
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
| | - Chongzhao Ran
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 Thirteenth street, Charlestown, Massachusetts 02129, United States
| | - Peter Caravan
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 Thirteenth street, Charlestown, Massachusetts 02129, United States
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5206, Boston, Massachusetts 02114, United States
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
31
|
Wen H, Cui Q, Meng H, Lai F, Wang S, Zhang X, Chen X, Cui H, Yin D. A high-resolution method to assess cell multinucleation with cytoplasm-localized fluorescent probes. Analyst 2016; 141:4010-3. [DOI: 10.1039/c6an00613b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell multinucleation is closely related to chromosomal instability.
Collapse
Affiliation(s)
- Hui Wen
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Qinghua Cui
- College of Pharmacy
- Shandong University of Traditional Chinese Medicine
- Jinan
- China
| | - Hui Meng
- College of Pharmacy
- Shandong University of Traditional Chinese Medicine
- Jinan
- China
| | - Fangfang Lai
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Shufang Wang
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Xiang Zhang
- Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Huaqing Cui
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine
- Institute of Materia Medica
- Peking Union Medical College and Chinese Academy of Medical Sciences
- Beijing
- China
| |
Collapse
|
32
|
A Metabolic Probe-Enabled Strategy Reveals Uptake and Protein Targets of Polyunsaturated Aldehydes in the Diatom Phaeodactylum tricornutum. PLoS One 2015; 10:e0140927. [PMID: 26496085 PMCID: PMC4619725 DOI: 10.1371/journal.pone.0140927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/01/2015] [Indexed: 01/12/2023] Open
Abstract
Diatoms are unicellular algae of crucial importance as they belong to the main primary producers in aquatic ecosystems. Several diatom species produce polyunsaturated aldehydes (PUAs) that have been made responsible for chemically mediated interactions in the plankton. PUA-effects include chemical defense by reducing the reproductive success of grazing copepods, allelochemical activity by interfering with the growth of competing phytoplankton and cell to cell signaling. We applied a PUA-derived molecular probe, based on the biologically highly active 2,4-decadienal, with the aim to reveal protein targets of PUAs and affected metabolic pathways. By using fluorescence microscopy, we observed a substantial uptake of the PUA probe into cells of the diatom Phaeodactylum tricornutum in comparison to the uptake of a structurally closely related control probe based on a saturated aldehyde. The specific uptake motivated a chemoproteomic approach to generate a qualitative inventory of proteins covalently targeted by the α,β,γ,δ-unsaturated aldehyde structure element. Activity-based protein profiling revealed selective covalent modification of target proteins by the PUA probe. Analysis of the labeled proteins gave insights into putative affected molecular functions and biological processes such as photosynthesis including ATP generation and catalytic activity in the Calvin cycle or the pentose phosphate pathway. The mechanism of action of PUAs involves covalent reactions with proteins that may result in protein dysfunction and interference of involved pathways.
Collapse
|
33
|
Murrey HE, Judkins JC, Am Ende CW, Ballard TE, Fang Y, Riccardi K, Di L, Guilmette ER, Schwartz JW, Fox JM, Johnson DS. Systematic Evaluation of Bioorthogonal Reactions in Live Cells with Clickable HaloTag Ligands: Implications for Intracellular Imaging. J Am Chem Soc 2015; 137:11461-75. [PMID: 26270632 PMCID: PMC4572613 DOI: 10.1021/jacs.5b06847] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Bioorthogonal
reactions, including the strain-promoted azide–alkyne
cycloaddition (SPAAC) and inverse electron demand Diels–Alder
(iEDDA) reactions, have become increasingly popular for live-cell
imaging applications. However, the stability and reactivity of reagents
has never been systematically explored in the context of a living
cell. Here we report a universal, organelle-targetable system based
on HaloTag protein technology for directly comparing bioorthogonal
reagent reactivity, specificity, and stability using clickable HaloTag
ligands in various subcellular compartments. This system enabled a
detailed comparison of the bioorthogonal reactions in live cells and
informed the selection of optimal reagents and conditions for live-cell
imaging studies. We found that the reaction of sTCO with monosubstituted
tetrazines is the fastest reaction in cells; however, both reagents
have stability issues. To address this, we introduced a new variant
of sTCO, Ag-sTCO, which has much improved stability and can be used
directly in cells for rapid bioorthogonal reactions with tetrazines.
Utilization of Ag complexes of conformationally strained trans-cyclooctenes should greatly expand their usefulness especially when
paired with less reactive, more stable tetrazines.
Collapse
Affiliation(s)
- Heather E Murrey
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| | - Joshua C Judkins
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| | - Christopher W Am Ende
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| | - T Eric Ballard
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States.,Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development , Groton, Connecticut 06340, United States
| | - Yinzhi Fang
- Brown Laboratories, Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware 19716, United States
| | - Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development , Groton, Connecticut 06340, United States
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development , Groton, Connecticut 06340, United States
| | - Edward R Guilmette
- Neuroscience and Pain Research Unit, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| | - Joel W Schwartz
- Neuroscience and Pain Research Unit, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| | - Joseph M Fox
- Brown Laboratories, Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware 19716, United States
| | - Douglas S Johnson
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
34
|
Tang TSM, Yip AMH, Zhang KY, Liu HW, Wu PL, Li KF, Cheah KW, Lo KKW. Bioorthogonal Labeling, Bioimaging, and Photocytotoxicity Studies of Phosphorescent Ruthenium(II) Polypyridine Dibenzocyclooctyne Complexes. Chemistry 2015; 21:10729-40. [DOI: 10.1002/chem.201501040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Indexed: 12/12/2022]
|
35
|
Vinegoni C, Dubach JM, Thurber GM, Miller MA, Mazitschek R, Weissleder R. Advances in measuring single-cell pharmacology in vivo. Drug Discov Today 2015; 20:1087-92. [PMID: 26024776 DOI: 10.1016/j.drudis.2015.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/10/2015] [Accepted: 05/20/2015] [Indexed: 11/26/2022]
Abstract
Measuring key pharmacokinetic and pharmacodynamic parameters in vivo at the single cell level is likely to enhance drug discovery and development. In this review, we summarize recent advances in this field and highlight current and future capabilities.
Collapse
Affiliation(s)
- Claudio Vinegoni
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA.
| | - J Matthew Dubach
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Greg M Thurber
- Department of Chemical Engineering, Department of Biomedical Engineering, University of Michigan, 2300 Hayward Avenue, Ann Arbor, MI 48109, USA
| | - Miles A Miller
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Ralph Mazitschek
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
36
|
Albrecht M, Lippach A, Exner MP, Jerbi J, Springborg M, Budisa N, Wenz G. Site-specific conjugation of 8-ethynyl-BODIPY to a protein by [2 + 3] cycloaddition. Org Biomol Chem 2015; 13:6728-36. [DOI: 10.1039/c5ob00505a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We report a straightforward synthesis of 8-ethynyl-BODIPY derivatives and their potential as fluorescent labeling compounds using an alkyne–azide click chemistry approach.
Collapse
Affiliation(s)
- Marcel Albrecht
- Organic Macromolecular Chemistry
- Campus Saarbrücken C4.2
- Saarland University
- D-66123 Saarbrücken
- Germany
| | - Andreas Lippach
- Organic Macromolecular Chemistry
- Campus Saarbrücken C4.2
- Saarland University
- D-66123 Saarbrücken
- Germany
| | | | - Jihene Jerbi
- Physical and Theoretical Chemistry
- Campus Saarbrücken B2.2
- Saarland University
- D-66123 Saarbrücken
- Germany
| | - Michael Springborg
- Physical and Theoretical Chemistry
- Campus Saarbrücken B2.2
- Saarland University
- D-66123 Saarbrücken
- Germany
| | - Nediljko Budisa
- Department of Chemistry-Biocatalysis
- TU Berlin
- D-10623 Berlin
- Germany
| | - Gerhard Wenz
- Organic Macromolecular Chemistry
- Campus Saarbrücken C4.2
- Saarland University
- D-66123 Saarbrücken
- Germany
| |
Collapse
|
37
|
Wu D, Cheung S, Daly R, Burke H, Scanlan EM, O'Shea DF. Synthesis and Glycoconjugation of an Azido-BF2-Azadipyrromethene Near-Infrared Fluorochrome. European J Org Chem 2014. [DOI: 10.1002/ejoc.201402960] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
38
|
Tercel M, McManaway SP, Liyanage HDS, Pruijn FB. Preparation and properties of clickable amino analogues of the duocarmycins: factors that affect the efficiency of their fluorescent labelling of DNA. ChemMedChem 2014; 9:2193-206. [PMID: 25044224 DOI: 10.1002/cmdc.201402169] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Indexed: 12/31/2022]
Abstract
Herein we report the synthesis of three DNA-alkylating amino analogues of the duocarmycins that carry an alkyne functional group suitable for copper-catalysed click chemistry. The alkyne-containing substituents are connected via a side chain position which projects away from the minor groove, and have only a small effect on DNA alkylation and cytotoxicity. The efficiency of click reactions with fluorophore azides was studied using alkylated ctDNA by analysing the adenine adducts produced after thermal depurination. Click reactions "on DNA" were sensitive to steric effects (tether length to the alkyne) and, surprisingly, to the nature of the fluorophore azide. With the best combination of click partners and reagents, adducts could be detected in the nuclei of treated cells by microscopy or flow cytometry, provided that an appropriate detergent (Triton X-100 and not Tween 20) was used for permeabilisation. The method is sensitive enough to detect adducts at physiologically relevant concentrations, and could have application in the development of nitro analogues of the duocarmycins as hypoxia-activated anticancer prodrugs.
Collapse
Affiliation(s)
- Moana Tercel
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142 (New Zealand), Fax: (+64) 9373-7502.
| | | | | | | |
Collapse
|
39
|
Rajendran M, Yapici E, Miller LW. Lanthanide-based imaging of protein-protein interactions in live cells. Inorg Chem 2014; 53:1839-53. [PMID: 24144069 PMCID: PMC3944735 DOI: 10.1021/ic4018739] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In order to deduce the molecular mechanisms of biological function, it is necessary to monitor changes in the subcellular location, activation, and interaction of proteins within living cells in real time. Förster resonance energy-transfer (FRET)-based biosensors that incorporate genetically encoded, fluorescent proteins permit high spatial resolution imaging of protein-protein interactions or protein conformational dynamics. However, a nonspecific fluorescence background often obscures small FRET signal changes, and intensity-based biosensor measurements require careful interpretation and several control experiments. These problems can be overcome by using lanthanide [Tb(III) or Eu(III)] complexes as donors and green fluorescent protein (GFP) or other conventional fluorophores as acceptors. Essential features of this approach are the long-lifetime (approximately milliseconds) luminescence of Tb(III) complexes and time-gated luminescence microscopy. This allows pulsed excitation, followed by a brief delay, which eliminates nonspecific fluorescence before the detection of Tb(III)-to-GFP emission. The challenges of intracellular delivery, selective protein labeling, and time-gated imaging of lanthanide luminescence are presented, and recent efforts to investigate the cellular uptake of lanthanide probes are reviewed. Data are presented showing that conjugation to arginine-rich, cell-penetrating peptides (CPPs) can be used as a general strategy for the cellular delivery of membrane-impermeable lanthanide complexes. A heterodimer of a luminescent Tb(III) complex, Lumi4, linked to trimethoprim and conjugated to nonaarginine via a reducible disulfide linker rapidly (∼10 min) translocates into the cytoplasm of Maden Darby canine kidney cells from the culture medium. With this reagent, the intracellular interaction between GFP fused to FK506 binding protein 12 (GFP-FKBP12) and the rapamycin binding domain of mTOR fused to Escherichia coli dihydrofolate reductase (FRB-eDHFR) were imaged at high signal-to-noise ratio with fast (1-3 s) image acquisition using a time-gated luminescence microscope. The data reviewed and presented here show that lanthanide biosensors enable fast, sensitive, and technically simple imaging of protein-protein interactions in live cells.
Collapse
Affiliation(s)
- Megha Rajendran
- Department of Chemistry, University of Illinois at Chicago, 845 West Taylor Street, Chicago, IL 60607
| | - Engin Yapici
- Department of Chemistry, University of Illinois at Chicago, 845 West Taylor Street, Chicago, IL 60607
| | - Lawrence W. Miller
- Department of Chemistry, University of Illinois at Chicago, 845 West Taylor Street, Chicago, IL 60607
| |
Collapse
|
40
|
Ghosh B, Jones LH. Target validation using in-cell small molecule clickable imaging probes. MEDCHEMCOMM 2014. [DOI: 10.1039/c3md00277b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The application of click chemistry to the visualization of chemical probes in in-cell chemical biology experiments is reviewed and the influence this research has had on target validation and molecular mode of action studies is also highlighted.
Collapse
Affiliation(s)
| | - Lyn H. Jones
- Pfizer
- Chemical Biology Group
- BioTherapeutics Chemistry
- WorldWide Medicinal Chemistry
- Cambridge
| |
Collapse
|
41
|
Abdo M, Sun Z, Knapp S. Biohybrid -Se-S- coupling reactions of an amino acid derived seleninate. Molecules 2013; 18:1963-72. [PMID: 23381022 PMCID: PMC6270073 DOI: 10.3390/molecules18021963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 01/28/2013] [Accepted: 01/30/2013] [Indexed: 11/24/2022] Open
Abstract
We describe the synthesis of the N-(2-seleninatoethyl) amide of N-Boc-phenylalanine, serving here as a peptide model, and its reductive coupling reactions under mild conditions with unprotected thiouridine and glutathione. Selenosulfide products such as these comprise reversibly conjugated bio-components, and can potentially find uses as probes of biological function, such as enzyme inhibitors, delivery systems, or structural mimics.
Collapse
Affiliation(s)
| | | | - Spencer Knapp
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-732-445-2627; Fax: +1-732-445-5312
| |
Collapse
|
42
|
Wüstner D, Solanko LM, Lund FW, Sage D, Schroll HJ, Lomholt MA. Quantitative fluorescence loss in photobleaching for analysis of protein transport and aggregation. BMC Bioinformatics 2012; 13:296. [PMID: 23148417 PMCID: PMC3557157 DOI: 10.1186/1471-2105-13-296] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 10/31/2012] [Indexed: 12/23/2022] Open
Abstract
Background Fluorescence loss in photobleaching (FLIP) is a widely used imaging technique, which provides information about protein dynamics in various cellular regions. In FLIP, a small cellular region is repeatedly illuminated by an intense laser pulse, while images are taken with reduced laser power with a time lag between the bleaches. Despite its popularity, tools are lacking for quantitative analysis of FLIP experiments. Typically, the user defines regions of interest (ROIs) for further analysis which is subjective and does not allow for comparing different cells and experimental settings. Results We present two complementary methods to detect and quantify protein transport and aggregation in living cells from FLIP image series. In the first approach, a stretched exponential (StrExp) function is fitted to fluorescence loss (FL) inside and outside the bleached region. We show by reaction–diffusion simulations, that the StrExp function can describe both, binding/barrier–limited and diffusion-limited FL kinetics. By pixel-wise regression of that function to FL kinetics of enhanced green fluorescent protein (eGFP), we determined in a user-unbiased manner from which cellular regions eGFP can be replenished in the bleached area. Spatial variation in the parameters calculated from the StrExp function allow for detecting diffusion barriers for eGFP in the nucleus and cytoplasm of living cells. Polyglutamine (polyQ) disease proteins like mutant huntingtin (mtHtt) can form large aggregates called inclusion bodies (IB’s). The second method combines single particle tracking with multi-compartment modelling of FL kinetics in moving IB’s to determine exchange rates of eGFP-tagged mtHtt protein (eGFP-mtHtt) between aggregates and the cytoplasm. This method is self-calibrating since it relates the FL inside and outside the bleached regions. It makes it therefore possible to compare release kinetics of eGFP-mtHtt between different cells and experiments. Conclusions We present two complementary methods for quantitative analysis of FLIP experiments in living cells. They provide spatial maps of exchange dynamics and absolute binding parameters of fluorescent molecules to moving intracellular entities, respectively. Our methods should be of great value for quantitative studies of intracellular transport.
Collapse
Affiliation(s)
- Daniel Wüstner
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark.
| | | | | | | | | | | |
Collapse
|
43
|
Jiang L, Zeng Y, Zhou H, Qu JY, Yao S. Visualizing millisecond chaotic mixing dynamics in microdroplets: A direct comparison of experiment and simulation. BIOMICROFLUIDICS 2012; 6:12810-1281012. [PMID: 22662077 PMCID: PMC3365329 DOI: 10.1063/1.3673254] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/08/2011] [Indexed: 05/08/2023]
Abstract
In order to fully explore and utilize the advantages of droplet-based microfluidics, fast, sensitive, and quantitative measurements are indispensable for the diagnosis of biochemical reactions in microdroplets. Here, we report an optical detection technique using two-photon fluorescence lifetime imaging microscopy, with an aligning-summing and non-fitting division method, to depict two-dimensional (2D) maps of mixing dynamics by chaotic advection in microdroplets with high temporal and spatial resolution. The mixing patterns of two dye solutions inside droplets were quantitatively and accurately measured. The mixing efficiency in a serpentine droplet mixer was also quantified and compared with the simulation data. The mapped chaotic mixing dynamics agree well with the numerical simulation and theoretical prediction. This quantitative characterization is potentially applicable to the real-time kinetic study of biological and chemical reactions in droplet-based microfluidic systems.
Collapse
|
44
|
Prokup A, Hemphill J, Deiters A. DNA computation: a photochemically controlled AND gate. J Am Chem Soc 2012; 134:3810-5. [PMID: 22239155 DOI: 10.1021/ja210050s] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
DNA computation is an emerging field that enables the assembly of complex circuits based on defined DNA logic gates. DNA-based logic gates have previously been operated through purely chemical means, controlling logic operations through DNA strands or other biomolecules. Although gates can operate through this manner, it limits temporal and spatial control of DNA-based logic operations. A photochemically controlled AND gate was developed through the incorporation of caged thymidine nucleotides into a DNA-based logic gate. By using light as the logic inputs, both spatial control and temporal control were achieved. In addition, design rules for light-regulated DNA logic gates were derived. A step-response, which can be found in a controller, was demonstrated. Photochemical inputs close the gap between DNA computation and silicon-based electrical circuitry, since light waves can be directly converted into electrical output signals and vice versa. This connection is important for the further development of an interface between DNA logic gates and electronic devices, enabling the connection of biological systems with electrical circuits.
Collapse
Affiliation(s)
- Alex Prokup
- Department of Chemistry, North Carolina State University, Raleigh, North Carolina 27695-8204, USA
| | | | | |
Collapse
|
45
|
Pauff SM, Miller SC. Synthesis of near-IR fluorescent oxazine dyes with esterase-labile sulfonate esters. Org Lett 2011; 13:6196-9. [PMID: 22047733 DOI: 10.1021/ol202619f] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Near-IR oxazine dyes are reported that contain sulfonate esters which are rapidly cleaved by esterase activity to unmask highly polar anionic sulfonates. Strategies for the synthesis of these dyes included the development of milder dye condensation conditions with improved functional compatibility and the use of an alkyl halide that allows for the introduction of esterase-labile sulfonates without the need for sulfonation of the target molecule.
Collapse
Affiliation(s)
- Steven M Pauff
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | |
Collapse
|
46
|
Li X, Zhu R, Yu A, Zhao XS. Ultrafast Photoinduced Electron Transfer between Tetramethylrhodamine and Guanosine in Aqueous Solution. J Phys Chem B 2011; 115:6265-71. [DOI: 10.1021/jp200455b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xun Li
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Department of Chemical Biology, College of Chemistry and Molecular Engineering, and Biodynamic Optical Imaging Center, Peking University, Beijing 100871, People's Republic of China
| | - Ruixue Zhu
- Department of Chemistry, Renmin University of China, Beijing 100872, People's Republic of China
| | - Anchi Yu
- Department of Chemistry, Renmin University of China, Beijing 100872, People's Republic of China
| | - Xin Sheng Zhao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Department of Chemical Biology, College of Chemistry and Molecular Engineering, and Biodynamic Optical Imaging Center, Peking University, Beijing 100871, People's Republic of China
| |
Collapse
|
47
|
Rusha L, Miller SC. Design and application of esterase-labile sulfonate protecting groups. Chem Commun (Camb) 2011; 47:2038-40. [PMID: 21210039 DOI: 10.1039/c0cc04796a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three esterase-labile, but chemically-stable sulfonate protecting groups have been designed and synthesized. One of these sulfonate esters allowed the cytoplasmic delivery and unmasking of a sulfonated dye in live cells.
Collapse
Affiliation(s)
- Laert Rusha
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation St., Worcester, MA 01605, USA
| | | |
Collapse
|
48
|
Jones LH, Beal D, Selby MD, Everson O, Burslem GM, Dodd P, Millbank J, Tran TD, Wakenhut F, Graham EJS, Targett-Adams P. In-cell click labelling of small molecules to determine subcellular localisation. J Chem Biol 2010; 4:49-53. [PMID: 22096474 DOI: 10.1007/s12154-010-0047-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 11/01/2010] [Indexed: 11/30/2022] Open
Abstract
UNLABELLED Small molecule fluorometric boron dipyrromethene probes were developed to bind hepatitis C virus-encoded NS5A protein and aid subcellular distribution studies. These molecules did not co-locate with NS5A, therefore alternative 'silent' azide reporters were used to obtain a more relevant picture of their distribution. Following pre-incubation with replicon cells, click chemistry was used to append a fluorophore to the azide that confirmed the co-localisation of the small molecule with the NS5A protein, thus providing greater insight into the antiviral mode of action of this chemotype. ELECTRONIC SUPPLEMENTARY MATERIAL The online version of this article (doi:10.1007/s12154-010-0047-1) contains supplementary material, which is available to authorized users.
Collapse
|