1
|
De Faveri C, Mattheisen JM, Sakmar TP, Coin I. Noncanonical Amino Acid Tools and Their Application to Membrane Protein Studies. Chem Rev 2024; 124:12498-12550. [PMID: 39509680 PMCID: PMC11613316 DOI: 10.1021/acs.chemrev.4c00181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024]
Abstract
Methods rooted in chemical biology have contributed significantly to studies of integral membrane proteins. One recent key approach has been the application of genetic code expansion (GCE), which enables the site-specific incorporation of noncanonical amino acids (ncAAs) with defined chemical properties into proteins. Efficient GCE is challenging, especially for membrane proteins, which have specialized biogenesis and cell trafficking machinery and tend to be expressed at low levels in cell membranes. Many eukaryotic membrane proteins cannot be expressed functionally in E. coli and are most effectively studied in mammalian cell culture systems. Recent advances have facilitated broader applications of GCE for studies of membrane proteins. First, AARS/tRNA pairs have been engineered to function efficiently in mammalian cells. Second, bioorthogonal chemical reactions, including cell-friendly copper-free "click" chemistry, have enabled linkage of small-molecule probes such as fluorophores to membrane proteins in live cells. Finally, in concert with advances in GCE methodology, the variety of available ncAAs has increased dramatically, thus enabling the investigation of protein structure and dynamics by multidisciplinary biochemical and biophysical approaches. These developments are reviewed in the historical framework of the development of GCE technology with a focus on applications to studies of membrane proteins.
Collapse
Affiliation(s)
- Chiara De Faveri
- Faculty
of Life Science, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| | - Jordan M. Mattheisen
- Laboratory
of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065, United States
- Tri-Institutional
PhD Program in Chemical Biology, New York, New York 10065, United States
| | - Thomas P. Sakmar
- Laboratory
of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065, United States
| | - Irene Coin
- Faculty
of Life Science, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
2
|
Jann C, Giofré S, Bhattacharjee R, Lemke EA. Cracking the Code: Reprogramming the Genetic Script in Prokaryotes and Eukaryotes to Harness the Power of Noncanonical Amino Acids. Chem Rev 2024; 124:10281-10362. [PMID: 39120726 PMCID: PMC11441406 DOI: 10.1021/acs.chemrev.3c00878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024]
Abstract
Over 500 natural and synthetic amino acids have been genetically encoded in the last two decades. Incorporating these noncanonical amino acids into proteins enables many powerful applications, ranging from basic research to biotechnology, materials science, and medicine. However, major challenges remain to unleash the full potential of genetic code expansion across disciplines. Here, we provide an overview of diverse genetic code expansion methodologies and systems and their final applications in prokaryotes and eukaryotes, represented by Escherichia coli and mammalian cells as the main workhorse model systems. We highlight the power of how new technologies can be first established in simple and then transferred to more complex systems. For example, whole-genome engineering provides an excellent platform in bacteria for enabling transcript-specific genetic code expansion without off-targets in the transcriptome. In contrast, the complexity of a eukaryotic cell poses challenges that require entirely new approaches, such as striving toward establishing novel base pairs or generating orthogonally translating organelles within living cells. We connect the milestones in expanding the genetic code of living cells for encoding novel chemical functionalities to the most recent scientific discoveries, from optimizing the physicochemical properties of noncanonical amino acids to the technological advancements for their in vivo incorporation. This journey offers a glimpse into the promising developments in the years to come.
Collapse
Affiliation(s)
- Cosimo Jann
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
Postdoc Programme (IPPro), 55128 Mainz, Germany
| | - Sabrina Giofré
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
Postdoc Programme (IPPro), 55128 Mainz, Germany
| | - Rajanya Bhattacharjee
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- IMB
International PhD Programme (IPP), 55128 Mainz, Germany
| | - Edward A. Lemke
- Biocenter, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Institute
of Molecular Biology (IMB), 55128 Mainz, Germany
| |
Collapse
|
3
|
Korovesis D, Gaspar VP, Beard HA, Chen S, Zahédi RP, Verhelst SHL. Mapping Peptide-Protein Interactions by Amine-Reactive Cleavable Photoaffinity Reagents. ACS OMEGA 2023; 8:25487-25495. [PMID: 37483247 PMCID: PMC10357517 DOI: 10.1021/acsomega.3c03064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023]
Abstract
Photoaffinity labeling followed by tandem mass spectrometry is an often used strategy to identify protein targets of small-molecule drugs or drug candidates, which, under ideal conditions, enables the identification of the actual drug binding site. In the case of bioactive peptides, however, identifying the distinct binding site is hampered because of complex fragmentation patterns during tandem mass spectrometry. We here report the development and use of small cleavable photoaffinity reagents that allow functionalization of bioactive peptides for light-induced covalent binding to their protein targets. Upon cleavage of the covalently linked peptide drug, a chemical remnant of a defined mass remains on the bound amino acid, which is then used to unambiguously identify the drug binding site. Applying our approach to known peptide-drug/protein pairs with reported crystal structures, such as the calmodulin-melittin interaction, we were able to validate the identified binding sites based on structural models. Overall, our cleavable photoaffinity labeling strategy represents a powerful tool to enable the identification of protein targets and specific binding sites of a wide variety of bioactive peptides in the future.
Collapse
Affiliation(s)
- Dimitris Korovesis
- Laboratory
of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven−University of Leuven, Herestraat 49 Box 802, Leuven 3000, Belgium
| | - Vanessa P. Gaspar
- Segal
Cancer Proteomics Centre, Lady Davis Institute
for Medical Research and McGill University, Montreal, Quebec H3T 1E2, Canada
- Gerald
Bronfman Department of Oncology, McGill
University, Montreal, Quebec H4A 3T2, Canada
| | - Hester A. Beard
- Laboratory
of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven−University of Leuven, Herestraat 49 Box 802, Leuven 3000, Belgium
| | - Suyuan Chen
- AG
Chemical Proteomics, Leibniz Institute for Analytical Sciences ISAS,
e.V., Otto-Hahn-Str. 6b, Dortmund 44227, Germany
| | - René P. Zahédi
- Segal
Cancer Proteomics Centre, Lady Davis Institute
for Medical Research and McGill University, Montreal, Quebec H3T 1E2, Canada
- Manitoba
Centre for Proteomics and Systems Biology, Winnipeg, Manitoba R3E 3P4, Canada
- Department
of Internal Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0Z2, Canada
- Department
of Biochemistry and Medical Genetics, University
of Manitoba, Winnipeg, Manitoba R3E 3N4, Canada
- Cancer
Care Manitoba Research Institute, Winnipeg, Manitoba R3E
0V9, Canada
| | - Steven H. L. Verhelst
- Laboratory
of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven−University of Leuven, Herestraat 49 Box 802, Leuven 3000, Belgium
- AG
Chemical Proteomics, Leibniz Institute for Analytical Sciences ISAS,
e.V., Otto-Hahn-Str. 6b, Dortmund 44227, Germany
| |
Collapse
|
4
|
Kotliar IB, Ceraudo E, Kemelmakher-Liben K, Oren DA, Lorenzen E, Dodig-Crnković T, Horioka-Duplix M, Huber T, Schwenk JM, Sakmar TP. Itch receptor MRGPRX4 interacts with the receptor activity-modifying proteins. J Biol Chem 2023; 299:104664. [PMID: 37003505 PMCID: PMC10165273 DOI: 10.1016/j.jbc.2023.104664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Cholestatic itch is a severe and debilitating symptom in liver diseases with limited treatment options. The class A G protein-coupled receptor (GPCR) Mas-related GPCR subtype X4 (MRGPRX4) has been identified as a receptor for bile acids, which are potential cholestatic pruritogens. An increasing number of GPCRs have been shown to interact with receptor activity-modifying proteins (RAMPs), which can modulate different aspects of GPCR biology. Using a combination of multiplexed immunoassay and proximity ligation assay, we show that MRGPRX4 interacts with RAMPs. The interaction of MRGPRX4 with RAMP2, but not RAMP1 or 3, causes attenuation of basal and agonist-dependent signaling, which correlates with a decrease of MRGPRX4 cell surface expression as measured using a quantitative NanoBRET pulse-chase assay. Finally, we use AlphaFold Multimer to predict the structure of the MRGPRX4-RAMP2 complex. The discovery that RAMP2 regulates MRGPRX4 may have direct implications for future drug development for cholestatic itch.
Collapse
Affiliation(s)
- Ilana B Kotliar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA; Tri-Institutional PhD Program in Chemical Biology, New York, New York, USA
| | - Emilie Ceraudo
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | - Kevin Kemelmakher-Liben
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | - Deena A Oren
- Structural Biology Resource Center, The Rockefeller University, New York, New York, USA
| | - Emily Lorenzen
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | - Tea Dodig-Crnković
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
| | - Mizuho Horioka-Duplix
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA
| | - Jochen M Schwenk
- Science for Life Laboratory, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Solna, Sweden
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York, USA; Department of Neurobiology, Care Sciences and Society, Section for Neurogeriatrics, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
5
|
Mattheisen JM, Wollowitz JS, Huber T, Sakmar TP. Genetic code expansion to enable site-specific bioorthogonal labeling of functional G protein-coupled receptors in live cells. Protein Sci 2023; 32:e4550. [PMID: 36540928 PMCID: PMC9847076 DOI: 10.1002/pro.4550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
For use in site-specific bioorthogonal labeling of expressed G protein-coupled receptors (GPCRs) in live cells, we developed a luciferase-based reporter assay. The assay was used to compare amber codon suppression efficiency, receptor functionality, and efficiency of different bioorthogonal labeling chemistries. We used the assay system to compare side-by-side the efficiency of incorporation of three different noncanonical amino acids [4-azido-l-phenylalanine (azF), cyclopropene-l-lysine (CpK), and trans-cyclooct-2-en-l-lysine (TCOK)] at three different sites on a GPCR using three different genetic code expansion plasmid systems. As a model GPCR, we engineered an epitope-tagged C-C chemokine receptor 5 (CCR5)-RLuc3 fusion for expression in HEK293T cells. Satisfactory incorporation of azF, CpK, and TCOK into heterologously expressed CCR5 was achieved. We also carried out cell-based calcium mobilization assays to measure the function of the engineered CCR5, and in the same cells, we performed bioorthogonal labeling of the engineered mutants using heterobivalent compounds containing bioorthogonal tethering groups linked to either a small-molecule fluorophore or a peptide. Favorable reaction kinetics of tetrazine-containing compounds with CCR5 harboring TCOK was observed. However, bioorthogonal labeling in live cells of CCR5 harboring CpK with tetrazine-containing compounds using the inverse electron demand Diels-Alder ligation was overall slightly more efficient than other reactions tested.
Collapse
Affiliation(s)
- Jordan M. Mattheisen
- Laboratory of Chemical Biology and Signal TransductionThe Rockefeller UniversityNew YorkNew YorkUSA
- Tri‐Institutional PhD Program in Chemical BiologyNew YorkNew YorkUSA
| | - Jaina S. Wollowitz
- Laboratory of Chemical Biology and Signal TransductionThe Rockefeller UniversityNew YorkNew YorkUSA
- Tri‐Institutional PhD Program in Chemical BiologyNew YorkNew YorkUSA
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal TransductionThe Rockefeller UniversityNew YorkNew YorkUSA
| | - Thomas P. Sakmar
- Laboratory of Chemical Biology and Signal TransductionThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
6
|
Yadav Y, Barman S, Roy A, Padhan J, Sudhamalla B. Uncovering the Domain-Specific Interactome of the TAF1 Tandem Reader Using Site-Specific Azide-Acetyllysine Photochemistry. Biochemistry 2023; 62:270-280. [PMID: 35786907 DOI: 10.1021/acs.biochem.2c00140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Combinatorial readout of histone post-translational modifications by tandem reader modules mediates crosstalk among different histone modifications. To identify the domain-specific interactome of the tandem reader, we engineered the dual bromodomain of TATA-binding protein-associated factor-1 (TAF1) to carry a photoactivatable unnatural amino acid, 4-azido-l-phenylalanine (AzF), via amber suppressor mutagenesis. Using computational approaches, we modeled the targeted residues of TAF1 with AzF to predict the cross-linking distance between the reactive arylazide and its interacting partner. We developed three photoactivatable TAF1 tandem-bromodomain analogues, viz., Y1403AzF in bromodomain 1 (BD1), W1526AzF in bromodomain 2 (BD2), and Y1403AzF/W1526AzF in both BD1 and BD2. Circular dichroism and a thermal shift assay were used to confirm the structural integrity of the engineered readers. Using the TAF1 tandem-bromodomain analogues, we characterized their histone ligand binding properties by isothermal titration calorimetry and photo-cross-linking experiments. We found that the dual bromodomain of TAF1 independently binds and cross-links to different acetylated histone ligands. We further used the engineered BD1 and BD2 analogues of the TAF1 tandem readers to identify their domain-specific interacting partners at the cellular level. Both BD1 and BD2 independently cross-link to a unique interactome, and importantly, the dual cross-linker carrying TAF1 analogue could capture both BD1- and BD2-specific interactomes. Our work concludes that BD1 and BD2 of the TAF1 tandem reader independently recognize their interacting partners to regulate downstream cellular functions.
Collapse
Affiliation(s)
- Yogita Yadav
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur, 741246 Nadia, West Bengal, India
| | - Soumen Barman
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur, 741246 Nadia, West Bengal, India
| | - Anirban Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur, 741246 Nadia, West Bengal, India
| | - Jyotirmayee Padhan
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur, 741246 Nadia, West Bengal, India
| | - Babu Sudhamalla
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur Campus, Mohanpur, 741246 Nadia, West Bengal, India
| |
Collapse
|
7
|
Kotliar IB, Lorenzen E, Schwenk JM, Hay DL, Sakmar TP. Elucidating the Interactome of G Protein-Coupled Receptors and Receptor Activity-Modifying Proteins. Pharmacol Rev 2023; 75:1-34. [PMID: 36757898 PMCID: PMC9832379 DOI: 10.1124/pharmrev.120.000180] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 09/27/2022] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are known to interact with several other classes of integral membrane proteins that modulate their biology and pharmacology. However, the extent of these interactions and the mechanisms of their effects are not well understood. For example, one class of GPCR-interacting proteins, receptor activity-modifying proteins (RAMPs), comprise three related and ubiquitously expressed single-transmembrane span proteins. The RAMP family was discovered more than two decades ago, and since then GPCR-RAMP interactions and their functional consequences on receptor trafficking and ligand selectivity have been documented for several secretin (class B) GPCRs, most notably the calcitonin receptor-like receptor. Recent bioinformatics and multiplexed experimental studies suggest that GPCR-RAMP interactions might be much more widespread than previously anticipated. Recently, cryo-electron microscopy has provided high-resolution structures of GPCR-RAMP-ligand complexes, and drugs have been developed that target GPCR-RAMP complexes. In this review, we provide a summary of recent advances in techniques that allow the discovery of GPCR-RAMP interactions and their functional consequences and highlight prospects for future advances. We also provide an up-to-date list of reported GPCR-RAMP interactions based on a review of the current literature. SIGNIFICANCE STATEMENT: Receptor activity-modifying proteins (RAMPs) have emerged as modulators of many aspects of G protein-coupled receptor (GPCR)biology and pharmacology. The application of new methodologies to study membrane protein-protein interactions suggests that RAMPs interact with many more GPCRs than had been previously known. These findings, especially when combined with structural studies of membrane protein complexes, have significant implications for advancing GPCR-targeted drug discovery and the understanding of GPCR pharmacology, biology, and regulation.
Collapse
Affiliation(s)
- Ilana B Kotliar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Emily Lorenzen
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Jochen M Schwenk
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Debbie L Hay
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York (I.B.K., E.L., T.P.S.); Tri-Institutional PhD Program in Chemical Biology, New York, New York (I.B.K.); Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Solna, Sweden (J.M.S.); Department of Pharmacology and Toxicology, School of Biomedical Sciences, Division of Health Sciences, University of Otago, Dunedin, New Zealand (D.L.H.); and Department of Neurobiology, Care Sciences and Society (NVS), Division for Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden (T.P.S.)
| |
Collapse
|
8
|
Leivaditis V, Koletsis E, Tsopanoglou N, Charokopos N, D’Alessandro C, Grapatsas K, Apostolakis E, Choleva E, Plota M, Emmanuil A, Dahm M, Dougenis D. The Coadministration of Levosimendan and Exenatide Offers a Significant Cardioprotective Effect to Isolated Rat Hearts against Ischemia/Reperfusion Injury. J Cardiovasc Dev Dis 2022; 9:jcdd9080263. [PMID: 36005427 PMCID: PMC9409795 DOI: 10.3390/jcdd9080263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
(1) Background: The present study aims to investigate the effect of administration of Levosimendan and Exenatide in various concentrations, as well as of the coadministration of those agents in an ischemia–reperfusion injury isolated heart model. (2) Methods: After 30 min of perfusion, the hearts underwent a 30 min period of regional ischemia followed by a 120 min period of reperfusion. All animals were randomly divided into 12 experimental groups of nine animals in each group: (1) Control, (2) Sham, (3) Digox (Negative control, Digoxin 1.67 μg/min), (4) Levo 1 (Levosimendan 0.01 μg/min), (5) Levo 2 (Levosimendan 0.03 μg/mL), (6) Levo 3 (Levosimendan 0.1 μg/min), (7) Levo 4 (Levosimendan 0.3 μg/min), (8) Levo 5 (Levosimendan 1 μg/min), (9) Exen 1 (Exenatide 0.001 μg/min), (10) Exen 2 (Exenatide 0.01 μg/min), (11) Exen 3 (Exenatide 0.1 μg/min) and (12) Combi (Levosimendan 0.1 µg/mL + Exenatide 0.001 μg/min). The hemodynamic parameters were recorded throughout the experiment. Arrhythmias and coronary flow were also evaluated. After every experiment the heart was suitably prepared and infarct size was measured. Markers of myocardial injury were also measured. Finally, oxidative stress was evaluated measuring reactive oxygen species. (3) Results: A dose-dependent improvement of the haemodynamic response was observed after the administration of both Levosimendan and Exenatide. The coadministration of both agents presented an even greater effect, improving the haemodynamic parameters further than the two agents separately. Levosimendan offered an increase of the coronary flow and both agents offered a reduction of arrhythmias. A dose-dependent reduction of the size of myocardial infarction and myocardial injury was observed after administration of Levosimendan and Exenatide. The coadministration of both agents offered a further improving the above parameters. Levosimendan also offered a significant reduction of oxidative stress. (4) Conclusions: The administration of Levosimendan and Exenatide offers a significant benefit by improving the haemodynamic response, increasing the coronary flow and reducing the occurrence of arrhythmias, the size of myocardial injury and myocardial oxidative stress in isolated rat hearts.
Collapse
Affiliation(s)
- Vasileios Leivaditis
- Department of Cardiothoracic and Vascular Surgery, Westpfalz-Klinikum, Hellmut-Hartert-Strasse 1, 67655 Kaiserslautern, Germany
- Department of Cardiothoracic Surgery, University Hospital of Patras, 26504 Patras, Greece
- Correspondence: ; Tel.: +49-151-50225145
| | - Efstratios Koletsis
- Department of Cardiothoracic Surgery, University Hospital of Patras, 26504 Patras, Greece
| | - Nikolaos Tsopanoglou
- Department of Pharmacology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Nikolaos Charokopos
- Department of Cardiothoracic Surgery, University Hospital of Patras, 26504 Patras, Greece
| | - Cristian D’Alessandro
- Laboratory of Biomechanics & Biomedical Engineering, Department of Mechanical Engineering & Aeronautics, University of Patras, 26504 Patras, Greece
| | - Konstantinos Grapatsas
- Department of Thoracic Surgery, Medical Center-University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| | - Efstratios Apostolakis
- Department of Cardiothoracic Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Effrosyni Choleva
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, 26504 Patras, Greece
| | - Maria Plota
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Andreas Emmanuil
- Laboratory of Hematology, University Hospital of Patras, 26504 Patras, Greece
| | - Manfred Dahm
- Department of Cardiothoracic and Vascular Surgery, Westpfalz-Klinikum, Hellmut-Hartert-Strasse 1, 67655 Kaiserslautern, Germany
| | - Dimitrios Dougenis
- Department of Cardiothoracic Surgery, Attikon University Hospital of Athens, 12462 Athens, Greece
| |
Collapse
|
9
|
Rudolf S, Kaempf K, Vu O, Meiler J, Beck‐Sickinger AG, Coin I. Binding of Natural Peptide Ligands to the Neuropeptide Y
5
Receptor. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202108738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Sarina Rudolf
- Institute of Biochemistry Faculty of Life Science Leipzig University 04103 Leipzig Germany
| | - Kerstin Kaempf
- Institute of Biochemistry Faculty of Life Science Leipzig University 04103 Leipzig Germany
| | - Oanh Vu
- Chemistry Department Vanderbilt University Nashville TN 37212 USA
| | - Jens Meiler
- Chemistry Department Vanderbilt University Nashville TN 37212 USA
- Center for Structural Biology Department of Biological Sciences Vanderbilt University Nashville TN 37212 USA
- Institute of Drug Design Faculty of Medicine Leipzig University 04103 Leipzig Germany
| | | | - Irene Coin
- Institute of Biochemistry Faculty of Life Science Leipzig University 04103 Leipzig Germany
| |
Collapse
|
10
|
Rudolf S, Kaempf K, Vu O, Meiler J, Beck-Sickinger AG, Coin I. Binding of Natural Peptide Ligands to the Neuropeptide Y 5 Receptor. Angew Chem Int Ed Engl 2022; 61:e202108738. [PMID: 34822209 PMCID: PMC8766924 DOI: 10.1002/anie.202108738] [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: 07/19/2021] [Indexed: 01/28/2023]
Abstract
The binding mode of natural peptide ligands to the Y5 G protein-coupled receptor (Y5 R), an attractive therapeutic target for the treatment of obesity, is largely unknown. Here, we apply complementary biochemical and computational approaches, including scanning of the receptor surface with a genetically encoded crosslinker, Ala-scanning of the ligand and double-cycle mutagenesis, to map interactions in the ligand-receptor interface and build a structural model of the NPY-Y5 R complex guided by the experimental data. In the model, the carboxyl (C)-terminus of bound NPY is placed close to the extracellular loop (ECL) 3, whereas the characteristic α-helical segment of the ligand drapes over ECL1 and is tethered towards ECL2 by a hydrophobic cluster. We further show that the other two natural ligands of Y5 R, peptide YY (PYY) and pancreatic polypeptide (PP) dock to the receptor in a similar pose.
Collapse
Affiliation(s)
- Sarina Rudolf
- Institute of Biochemistry, Faculty of Life Science, Leipzig University, Leipzig 04103, Germany
| | - Kerstin Kaempf
- Institute of Biochemistry, Faculty of Life Science, Leipzig University, Leipzig 04103, Germany
| | - Oanh Vu
- Chemistry Department, Vanderbilt University, Nashville, Tennessee 37212, U.S.A
| | - Jens Meiler
- Chemistry Department, Vanderbilt University, Nashville, Tennessee 37212, U.S.A
- Center for Structural Biology, Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37212, U.S.A
- Institute of Drug Design, Faculty of Medicine, Leipzig University, Leipzig 04103, Germany
| | | | - Irene Coin
- Institute of Biochemistry, Faculty of Life Science, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
11
|
Lucey M, Ashik T, Marzook A, Wang Y, Goulding J, Oishi A, Broichhagen J, Hodson DJ, Minnion J, Elani Y, Jockers R, Briddon SJ, Bloom SR, Tomas A, Jones B. Acylation of the Incretin Peptide Exendin-4 Directly Impacts Glucagon-Like Peptide-1 Receptor Signaling and Trafficking. Mol Pharmacol 2021; 100:319-334. [PMID: 34315812 PMCID: PMC8626645 DOI: 10.1124/molpharm.121.000270] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022] Open
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a class B G protein-coupled receptor and mainstay therapeutic target for the treatment of type 2 diabetes and obesity. Recent reports have highlighted how biased agonism at the GLP-1R affects sustained glucose-stimulated insulin secretion through avoidance of desensitization and downregulation. A number of GLP-1R agonists (GLP-1RAs) feature a fatty acid moiety to prolong their pharmacokinetics via increased albumin binding, but the potential for these chemical changes to influence GLP-1R function has rarely been investigated beyond potency assessments for cAMP. Here, we directly compare the prototypical GLP-1RA exendin-4 with its C-terminally acylated analog, exendin-4-C16. We examine relative propensities of each ligand to recruit and activate G proteins and β-arrestins, endocytic and postendocytic trafficking profiles, and interactions with model and cellular membranes in HEK293 and HEK293T cells. Both ligands had similar cAMP potency, but exendin-4-C16 showed ∼2.5-fold bias toward G protein recruitment and a ∼60% reduction in β-arrestin-2 recruitment efficacy compared with exendin-4, as well as reduced GLP-1R endocytosis and preferential targeting toward recycling pathways. These effects were associated with reduced movement of the GLP-1R extracellular domain measured using a conformational biosensor approach and a ∼70% increase in insulin secretion in INS-1 832/3 cells. Interactions with plasma membrane lipids were enhanced by the acyl chain. Exendin-4-C16 showed extensive albumin binding and was highly effective for lowering of blood glucose in mice over at least 72 hours. Our study highlights the importance of a broad approach to the evaluation of GLP-1RA pharmacology. SIGNIFICANCE STATEMENT: Acylation is a common strategy to enhance the pharmacokinetics of peptide-based drugs. This work shows how acylation can also affect various other pharmacological parameters, including biased agonism, receptor trafficking, and interactions with the plasma membrane, which may be therapeutically important.
Collapse
Affiliation(s)
- Maria Lucey
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Tanyel Ashik
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Amaara Marzook
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Yifan Wang
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Joëlle Goulding
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Atsuro Oishi
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Johannes Broichhagen
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - David J Hodson
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - James Minnion
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Yuval Elani
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Ralf Jockers
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Stephen J Briddon
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Stephen R Bloom
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Alejandra Tomas
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine (M.L., T.A., A.M., J.M., S.R.B., B.J.) and Section of Cell Biology and Functional Genomics (Y.W., A.T.), Department of Metabolism, Digestion and Reproduction, and Department of Chemical Engineering (Y.E.), Imperial College London, London, United Kingdom; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (J.G., S.J.B.); Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, United Kingdom (J.G., D.J.H., S.J.B.); Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France (A.O., R.J.); Department of Anatomy, Kyorin University Faculty of Medicine, Tokyo, Japan (A.O.); Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (J.B.); Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, United Kingdom (D.J.H.); and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom (D.J.H.)
| |
Collapse
|
12
|
Capturing Peptide-GPCR Interactions and Their Dynamics. Molecules 2020; 25:molecules25204724. [PMID: 33076289 PMCID: PMC7587574 DOI: 10.3390/molecules25204724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Many biological functions of peptides are mediated through G protein-coupled receptors (GPCRs). Upon ligand binding, GPCRs undergo conformational changes that facilitate the binding and activation of multiple effectors. GPCRs regulate nearly all physiological processes and are a favorite pharmacological target. In particular, drugs are sought after that elicit the recruitment of selected effectors only (biased ligands). Understanding how ligands bind to GPCRs and which conformational changes they induce is a fundamental step toward the development of more efficient and specific drugs. Moreover, it is emerging that the dynamic of the ligand–receptor interaction contributes to the specificity of both ligand recognition and effector recruitment, an aspect that is missing in structural snapshots from crystallography. We describe here biochemical and biophysical techniques to address ligand–receptor interactions in their structural and dynamic aspects, which include mutagenesis, crosslinking, spectroscopic techniques, and mass-spectrometry profiling. With a main focus on peptide receptors, we present methods to unveil the ligand–receptor contact interface and methods that address conformational changes both in the ligand and the GPCR. The presented studies highlight a wide structural heterogeneity among peptide receptors, reveal distinct structural changes occurring during ligand binding and a surprisingly high dynamics of the ligand–GPCR complexes.
Collapse
|
13
|
Naider F, Becker JM. A Paradigm for Peptide Hormone-GPCR Analyses. Molecules 2020; 25:E4272. [PMID: 32961885 PMCID: PMC7570734 DOI: 10.3390/molecules25184272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 01/14/2023] Open
Abstract
Work from our laboratories over the last 35 years that has focused on Ste2p, a G protein-coupled receptor (GPCR), and its tridecapeptide ligand α-factor is reviewed. Our work utilized the yeast Saccharomyces cerevisiae as a model system for understanding peptide-GPCR interactions. It explored the structure and function of synthetic α-factor analogs and biosynthetic receptor domains, as well as designed mutations of Ste2p. The results and conclusions are described using the nuclear magnetic resonance interrogation of synthetic Ste2p transmembrane domains (TMs), the fluorescence interrogation of agonist and antagonist binding, the biochemical crosslinking of peptide analogs to Ste2p, and the phenotypes of receptor mutants. We identified the ligand-binding domain in Ste2p, the functional assemblies of TMs, unexpected and interesting ligand analogs; gained insights into the bound α-factor structure; and unraveled the function and structures of various Ste2p domains, including the N-terminus, TMs, loops connecting the TMs, and the C-terminus. Our studies showed interactions between specific residues of Ste2p in an active state, but not resting state, and the effect of ligand activation on the dimerization of Ste2p. We show that, using a battery of different biochemical and genetic approaches, deep insight can be gained into the structure and conformational dynamics of GPCR-peptide interactions in the absence of a crystal structure.
Collapse
Affiliation(s)
- Fred Naider
- Department of Chemistry, College of Staten Island, CUNY, 2800 Victory Blvd, Staten Island, NY 10314, USA
| | - Jeffrey M. Becker
- Department of Microbiology, University of Tennessee, 610 Ken and Blaire Mossman Building, 1311 Cumberland Avenue, Knoxville, TN 37996, USA
| |
Collapse
|
14
|
Pickford P, Lucey M, Fang Z, Bitsi S, de la Serna JB, Broichhagen J, Hodson DJ, Minnion J, Rutter GA, Bloom SR, Tomas A, Jones B. Signalling, trafficking and glucoregulatory properties of glucagon-like peptide-1 receptor agonists exendin-4 and lixisenatide. Br J Pharmacol 2020; 177:3905-3923. [PMID: 32436216 PMCID: PMC7429481 DOI: 10.1111/bph.15134] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Amino acid substitutions at the N-termini of glucagon-like peptide-1 (GLP-1) receptor agonist peptides result in distinct patterns of intracellular signalling, sub-cellular trafficking and efficacy in vivo. Here, we to determine whether sequence differences at the ligand C-termini of clinically approved GLP-1 receptor agonists exendin-4 and lixisenatide lead to similar phenomena. EXPERIMENTAL APPROACH Exendin-4, lixisenatide and N-terminally substituted analogues with biased signalling characteristics were compared across a range of in vitro trafficking and signalling assays in different cell types. Fluorescent ligands and new time-resolved FRET approaches were developed to study agonist behaviours at the cellular and sub-cellular level. Anti-hyperglycaemic and anorectic effects of each parent ligand and their biased derivatives were assessed in mice. KEY RESULTS Lixisenatide and exendin-4 showed equal binding affinity, but lixisenatide was fivefold less potent for cAMP signalling. Both peptides induced extensive GLP-1 receptor clustering in the plasma membrane and were rapidly endocytosed, but the GLP-1 receptor recycled more slowly to the cell surface after lixisenatide treatment. These combined deficits resulted in reduced maximal sustained insulin secretion and reduced anti-hyperglycaemic and anorectic effects in mice with lixisenatide. N-terminal substitution of His1 by Phe1 to both ligands had favourable effects on their pharmacology, resulting in improved insulin release and lowering of blood glucose. CONCLUSION AND IMPLICATIONS Changes to the C-terminus of exendin-4 affect signalling potency and GLP-1 receptor trafficking via mechanisms unrelated to GLP-1 receptor occupancy. These differences were associated with changes in their ability to control blood glucose and therefore may be therapeutically relevant.
Collapse
Affiliation(s)
- Philip Pickford
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| | - Maria Lucey
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| | - Zijian Fang
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| | - Stavroula Bitsi
- Section of Cell Biology and Functional GenomicsImperial College LondonLondonUK
| | | | - Johannes Broichhagen
- Department Chemical BiologyMax Planck Institute for Medical ResearchHeidelbergGermany
- Department Chemical BiologyLeibniz‐Forschungsinstitut für Molekulare Pharmakologie (FMP)BerlinGermany
| | - David J. Hodson
- Institute of Metabolism and Systems Research (IMSR), and Centre of Membrane Proteins and Receptors (COMPARE)University of BirminghamBirminghamUK
- Centre for Endocrinology, Diabetes and MetabolismBirmingham Health PartnersBirminghamUK
| | - James Minnion
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| | - Guy A. Rutter
- Section of Cell Biology and Functional GenomicsImperial College LondonLondonUK
| | - Stephen R. Bloom
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| | - Alejandra Tomas
- Section of Cell Biology and Functional GenomicsImperial College LondonLondonUK
| | - Ben Jones
- Section of Endocrinology and Investigative MedicineImperial College LondonLondonUK
| |
Collapse
|
15
|
Kadunc L, Svetličič M, Forstnerič V, Hafner Bratkovič I, Jerala R. Increased gene translation stringency in mammalian cells by nonsense suppression at multiple permissive sites with a single noncanonical amino acid. FEBS Lett 2020; 594:2452-2461. [PMID: 32401336 DOI: 10.1002/1873-3468.13810] [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: 03/02/2020] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 11/10/2022]
Abstract
The considerable potential of engineered cells compels the development of strategies for the stringent control of gene expression. A promising approach is the introduction of a premature stop codon (PTC) into a selected gene that is expressed only in the presence of noncanonical amino acids through nonsense suppression. Here, different strategies of amber PTC readthrough in mammalian cells were tested. The use of a tRNA synthetase together with a TAG codon-specific tRNA achieved PTC readthrough depending on the addition of a noncanonical amino acid (4-benzoyl-L-phenylalanine; Bpa). While single TAG codon incorporation exhibited detectable expression of the reporter protein even in the absence of Bpa, the use of a double PTC enabled virtually leakage-free functional gene translation. The introduction of an additional 5'-PTC, therefore, represents a generally applicable strategy to increase stringency in gene translation.
Collapse
Affiliation(s)
- Lucija Kadunc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,Graduate School of Biomedicine, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Svetličič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Vida Forstnerič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Iva Hafner Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.,EN-FIST Centre of Excellence, Ljubljana, Slovenia
| |
Collapse
|
16
|
Fang Z, Chen S, Pickford P, Broichhagen J, Hodson DJ, Corrêa IR, Kumar S, Görlitz F, Dunsby C, French PMW, Rutter GA, Tan T, Bloom SR, Tomas A, Jones B. The Influence of Peptide Context on Signaling and Trafficking of Glucagon-like Peptide-1 Receptor Biased Agonists. ACS Pharmacol Transl Sci 2020; 3:345-360. [PMID: 32296773 PMCID: PMC7155199 DOI: 10.1021/acsptsci.0c00022] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Indexed: 01/14/2023]
Abstract
Signal bias and membrane trafficking have recently emerged as important considerations in the therapeutic targeting of the glucagon-like peptide-1 receptor (GLP-1R) in type 2 diabetes and obesity. In the present study, we have evaluated a peptide series with varying sequence homology between native GLP-1 and exendin-4, the archetypal ligands on which approved GLP-1R agonists are based. We find notable differences in agonist-mediated cyclic AMP signaling, recruitment of β-arrestins, endocytosis, and recycling, dependent both on the introduction of a His → Phe switch at position 1 and the specific midpeptide helical regions and C-termini of the two agonists. These observations were linked to insulin secretion in a beta cell model and provide insights into how ligand factors influence GLP-1R function at the cellular level.
Collapse
Affiliation(s)
- Zijian Fang
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Shiqian Chen
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Philip Pickford
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Johannes Broichhagen
- Department
Chemical Biology, Leibniz-Forschungsinstitut
für Molekulare Pharmakologie (FMP), Berlin, 13125, Germany
| | - David J. Hodson
- Institute
of Metabolism and Systems Research (IMSR), and Centre of Membrane
Proteins and Receptors (COMPARE), University
of Birmingham, Birmingham, B15 2TT, United Kingdom
- Centre
for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, B15 2TT, United Kingdom
| | - Ivan R. Corrêa
- New
England
Biolabs, Ipswich, Massachusetts 01938, United States
| | - Sunil Kumar
- Department
of Physics, Imperial College London, London, SW7 2BX, United Kingdom
| | - Frederik Görlitz
- Department
of Physics, Imperial College London, London, SW7 2BX, United Kingdom
| | - Chris Dunsby
- Department
of Physics, Imperial College London, London, SW7 2BX, United Kingdom
| | - Paul M. W. French
- Department
of Physics, Imperial College London, London, SW7 2BX, United Kingdom
| | - Guy A. Rutter
- Section
of Cell Biology and Functional Genomics, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Tricia Tan
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Stephen R. Bloom
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Alejandra Tomas
- Section
of Cell Biology and Functional Genomics, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Ben Jones
- Section
of Endocrinology and Investigative Medicine, Imperial College London, London, W12 0NN, United Kingdom
| |
Collapse
|
17
|
Dong M, Harikumar KG, Raval SR, Milburn JE, Clark C, Alcala-Torano R, Mobarec JC, Reynolds CA, Ghirlanda G, Christopoulos A, Wootten D, Sexton PM, Miller LJ. Rational development of a high-affinity secretin receptor antagonist. Biochem Pharmacol 2020; 177:113929. [PMID: 32217097 DOI: 10.1016/j.bcp.2020.113929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/19/2020] [Indexed: 01/11/2023]
Abstract
The secretin receptor is a prototypic class B GPCR with substantial and broad pharmacologic importance. The aim of this project was to develop a high affinity selective antagonist as a new and important pharmacologic tool and to aid stabilization of this receptor in an inactive conformation for ultimate structural characterization. Amino-terminal truncation of the natural 27-residue ligand reduced biological activity, but also markedly reduced binding affinity. This was rationally and experimentally overcome with lactam stabilization of helical structure and with replacement of residues with natural and unnatural amino acids. A key new step in this effort was the replacement of peptide residue Leu22 with L-cyclohexylalanine (Cha) to enhance potential hydrophobic interactions with receptor residues Leu31, Val34, and Phe92 that were predicted from molecular modeling. Alanine-replacement mutagenesis of these residues markedly affected ligand binding and biological activity. The optimal antagonist ligand, (Y10,c[E16,K20],I17,Cha22,R25)sec(6-27), exhibited high binding affinity (4 nM), similar to natural secretin, and exhibited no demonstrable biological activity to stimulate cAMP accumulation, intracellular calcium mobilization, or β-arrestin-2 translocation. It acts as an orthosteric competitive antagonist, predicted to bind within the peptide-binding groove in the receptor extracellular domain. The analogous peptide that was one residue longer, retaining Thr5, exhibited partial agonist activity, while further truncation of even a single residue (Phe6) reduced binding affinity. This sec(6-27)-based peptide will be an important new tool for pharmacological and structural studies.
Collapse
Affiliation(s)
- Maoqing Dong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, United States
| | - Kaleeckal G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, United States
| | - Sweta R Raval
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, United States
| | - Juliana E Milburn
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, United States
| | - Carolyn Clark
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, United States
| | - Rafael Alcala-Torano
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, United States
| | - Juan C Mobarec
- Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Christopher A Reynolds
- Department of Biological Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Giovanna Ghirlanda
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85281, United States
| | - Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 Victoria, Australia
| | - Denise Wootten
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 Victoria, Australia
| | - Patrick M Sexton
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052 Victoria, Australia
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, United States.
| |
Collapse
|
18
|
Giusti F, Casiraghi M, Point E, Damian M, Rieger J, Bon CL, Pozza A, Moncoq K, Banères JL, Catoire LJ. Structure of the agonist 12-HHT in its BLT2 receptor-bound state. Sci Rep 2020; 10:2630. [PMID: 32060341 PMCID: PMC7021728 DOI: 10.1038/s41598-020-59571-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/28/2020] [Indexed: 12/25/2022] Open
Abstract
G Protein-Coupled receptors represent the main communicating pathway for signals from the outside to the inside of most of eukaryotic cells. They define the largest family of integral membrane receptors at the surface of the cells and constitute the main target of the current drugs on the market. The low affinity leukotriene receptor BLT2 is a receptor involved in pro- and anti-inflammatory pathways and can be activated by various unsaturated fatty acid compounds. We present here the NMR structure of the agonist 12-HHT in its BLT2-bound state and a model of interaction of the ligand with the receptor based on a conformational homology modeling associated with docking simulations. Put into perspective with the data obtained with leukotriene B4, our results illuminate the ligand selectivity of BLT2 and may help define new molecules to modulate the activity of this receptor.
Collapse
Affiliation(s)
- Fabrice Giusti
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France.,Institut de Chimie Séparative de Marcoule, ICSM UMR 5257, Site de Marcoule, Bâtiment 426, BP 17171, F-30207, Bagnols sur Cèze Cedex, France
| | - Marina Casiraghi
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, 94305, Stanford California, USA
| | - Elodie Point
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France
| | - Marjorie Damian
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université Montpellier, ENSCM, , 15 av. Charles Flahault, 34093, Montpellier, France
| | - Jutta Rieger
- Institut Parisien de Chimie Moléculaire, Sorbonne Université, CNRS, UMR 8232, Equipe Chimie des Polymères, 4 place Jussieu, 75252, Paris Cedex, 05, France
| | - Christel Le Bon
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France
| | - Alexandre Pozza
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France
| | - Karine Moncoq
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS, Université Montpellier, ENSCM, , 15 av. Charles Flahault, 34093, Montpellier, France
| | - Laurent J Catoire
- Laboratoire de Biologie Physico-Chimique des Protéines Membranaires, UMR 7099, CNRS/Université de Paris, Institut de Biologie Physico-Chimique (FRC 550), 13 rue Pierre et Marie Curie, F-75005, Paris, France.
| |
Collapse
|
19
|
Song X, Yu Y, Shen C, Wang Y, Wang N. Dimerization/oligomerization of the extracellular domain of the GLP-1 receptor and the negative cooperativity in its ligand binding revealed by the improved NanoBiT. FASEB J 2020; 34:4348-4368. [PMID: 31970836 DOI: 10.1096/fj.201902007r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/15/2019] [Accepted: 01/12/2020] [Indexed: 12/13/2022]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R), a family B G-protein coupled receptor (GPCR), regulates the insulin secretion following stimulation by ligands. The transmembrane domain (TM) mediates GLP-1R homodimerization, which modulates its ligand binding and signaling. We investigated the possible involvement of the N-terminal extracellular domain (NTD) in dimerization/oligomerization and dimer-associated ligand binding by NanoLuc Binary Technology (NanoBiT). With improved NanoBiT detection using a decreasing substrate concentration, the negative cooperativity of ligand binding to the NTD was confirmed by accelerated dissociation and Scatchard analysis. The dimerization/oligomerization of the isolated NTD was observed by NanoBiT and validated by analytical ultracentrifugation, deriving the comparable dimerization affinity (~105 M-1 ). The NTD was also involved in the dimerization/oligomerization of the full-length GLP-1R with mutated TM4 at the cellular level. In an analysis of the parameters of the NTD binding, the Kd for the probe GLP-1 (7-36, A8G) was similar (6-8 μM) in both the 1:1 binding model and the receptor dimerization model. Compared with GLP-1 and dulaglutide, exenatide showed two-site binding with Ki values of 1.4 pM and 8.7 nM. Our study indicates the involvement of NTD in the GLP-1R dimerization/oligomerization and suggests that further investigations on the role in other family B GPCRs are needed.
Collapse
Affiliation(s)
- Xiaohan Song
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Yi Yu
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Cangjie Shen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Yubo Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Nan Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
20
|
Ablenas CJ, Gidi Y, Powdrill MH, Ahmed N, Shaw TA, Mesko M, Götte M, Cosa G, Pezacki JP. Hepatitis C Virus Helicase Binding Activity Monitored through Site-Specific Labeling Using an Expanded Genetic Code. ACS Infect Dis 2019; 5:2118-2126. [PMID: 31640339 DOI: 10.1021/acsinfecdis.9b00220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mechanism of unwinding catalyzed by the hepatitis C virus nonstructural protein 3 helicase (NS3h) has been a subject of considerable interest, with NS3h serving as a prototypical enzyme in the study of helicase function. Recent studies support an ATP-fueled, inchworm-like stepping of NS3h on the nucleic acid that would result in the displacement of the complementary strand of the duplex during unwinding. Here, we describe the screening of a site of incorporation of an unnatural amino acid in NS3h for fluorescent labeling of the enzyme to be used in single-molecule Förster resonance energy transfer (FRET) experiments. From the nine potential sites identified in NS3h for incorporation of the unnatural amino acid, only one allowed for expression and fluorescent labeling of the recombinant protein. Incorporation of the unnatural amino acid was confirmed via bulk assays to not interfere with unwinding activity of the helicase. Binding to four different dsDNA sequences bearing a ssDNA overhang segment of varying length (either minimal 6 or 7 base length overhang to ensure binding or a long 24 base overhang) and sequence was recorded with the new NS3h construct at the single-molecule level. Single-molecule fluorescence displayed time intervals with anticorrelated donor and acceptor emission fluctuations associated with protein binding to the substrates. An apparent FRET value was estimated from the binding events showing a single FRET value of ∼0.8 for the 6-7 base overhangs. A smaller mean value and a broad distribution was in turn recorded for the long ssDNA overhang, consistent with NS3h exploring a larger physical space while bound to the DNA construct. Notably, intervals where NS3h binding was recorded were exhibited at time periods where the acceptor dye reversibly bleached. Protein induced fluorescence intensity enhancement in the donor channel became apparent at these intervals. Overall, the site-specific fluorescent labeling of NS3h reported here provides a powerful tool for future studies to monitor the dynamics of enzyme translocation during unwinding by single-molecule FRET.
Collapse
Affiliation(s)
- Christopher J. Ablenas
- Department of Biochemistry, McGill University, Montreal, Quebec H3G1Y6, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| | - Yasser Gidi
- Department of Chemistry, McGill University, Montreal, Quebec H3A0B8, Canada
| | - Megan H. Powdrill
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| | - Noreen Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| | - Tyler A. Shaw
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| | - Mihai Mesko
- Department of Chemistry, McGill University, Montreal, Quebec H3A0B8, Canada
| | - Matthias Götte
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G2R7, Canada
| | - Gonzalo Cosa
- Department of Chemistry, McGill University, Montreal, Quebec H3A0B8, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N6N5, Canada
| |
Collapse
|
21
|
Gagnon L, Cao Y, Cho A, Sedki D, Huber T, Sakmar TP, Laporte SA. Genetic code expansion and photocross-linking identify different β-arrestin binding modes to the angiotensin II type 1 receptor. J Biol Chem 2019; 294:17409-17420. [PMID: 31530642 DOI: 10.1074/jbc.ra119.010324] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/05/2019] [Indexed: 12/28/2022] Open
Abstract
The angiotensin II (AngII) type 1 receptor (AT1R) is a member of the G protein-coupled receptor (GPCR) family and binds β-arrestins (β-arrs), which regulate AT1R signaling and trafficking. These processes can be biased by different ligands or mutations in the AGTR1 gene. As for many GPCRs, the exact details for AT1R-β-arr interactions driven by AngII or β-arr-biased ligands remain largely unknown. Here, we used the amber-suppression technology to site-specifically introduce the unnatural amino acid (UAA) p-azido-l-phenylalanine (azF) into the intracellular loops (ICLs) and the C-tail of AT1R. Our goal was to generate competent photoreactive receptors that can be cross-linked to β-arrs in cells. We performed UV-mediated photolysis of 25 different azF-labeled AT1Rs to cross-link β-arr1 to AngII-bound receptors, enabling us to map important contact sites in the C-tail and in the ICL2 and ICL3 of the receptor. The extent of AT1R-β-arr1 cross-linking among azF-labeled receptors differed, revealing variability in β-arr's contact mode with the different AT1R domains. Moreover, the signature of ligated AT1R-β-arr complexes from a subset of azF-labeled receptors also differed between AngII and β-arr-biased ligand stimulation of receptors and between azF-labeled AT1R bearing and that lacking a bias signaling mutation. These observations further implied distinct interaction modalities of the AT1R-β-arr1 complex in biased signaling conditions. Our findings demonstrate that this photocross-linking approach is useful for understanding GPCR-β-arr complexes in different activation states and could be extended to study other protein-protein interactions in cells.
Collapse
Affiliation(s)
- Laurence Gagnon
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Yubo Cao
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Aaron Cho
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Dana Sedki
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065
| | - Stéphane A Laporte
- Department of Medicine, Research Institute of the McGill University Health Center, McGill University, Montréal, Québec H4A 3J1, Canada .,Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
22
|
Tomas A, Jones B, Leech C. New Insights into Beta-Cell GLP-1 Receptor and cAMP Signaling. J Mol Biol 2019; 432:1347-1366. [PMID: 31446075 DOI: 10.1016/j.jmb.2019.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
Harnessing the translational potential of the GLP-1/GLP-1R system in pancreatic beta cells has led to the development of established GLP-1R-based therapies for the long-term preservation of beta cell function. In this review, we discuss recent advances in the current research on the GLP-1/GLP-1R system in beta cells, including the regulation of signaling by endocytic trafficking as well as the application of concepts such as signal bias, allosteric modulation, dual agonism, polymorphic receptor variants, spatial compartmentalization of cAMP signaling and new downstream signaling targets involved in the control of beta cell function.
Collapse
Affiliation(s)
- Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK.
| | - Ben Jones
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Colin Leech
- Department of Surgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| |
Collapse
|
23
|
Nödling AR, Spear LA, Williams TL, Luk LYP, Tsai YH. Using genetically incorporated unnatural amino acids to control protein functions in mammalian cells. Essays Biochem 2019; 63:237-266. [PMID: 31092687 PMCID: PMC6610526 DOI: 10.1042/ebc20180042] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
Genetic code expansion allows unnatural (non-canonical) amino acid incorporation into proteins of interest by repurposing the cellular translation machinery. The development of this technique has enabled site-specific incorporation of many structurally and chemically diverse amino acids, facilitating a plethora of applications, including protein imaging, engineering, mechanistic and structural investigations, and functional regulation. Particularly, genetic code expansion provides great tools to study mammalian proteins, of which dysregulations often have important implications in health. In recent years, a series of methods has been developed to modulate protein function through genetically incorporated unnatural amino acids. In this review, we will first discuss the basic concept of genetic code expansion and give an up-to-date list of amino acids that can be incorporated into proteins in mammalian cells. We then focus on the use of unnatural amino acids to activate, inhibit, or reversibly modulate protein function by translational, optical or chemical control. The features of each approach will also be highlighted.
Collapse
Affiliation(s)
| | - Luke A Spear
- School of Chemistry, Cardiff University, Cardiff, Wales, United Kingdom
| | - Thomas L Williams
- School of Chemistry, Cardiff University, Cardiff, Wales, United Kingdom
| | - Louis Y P Luk
- School of Chemistry, Cardiff University, Cardiff, Wales, United Kingdom
| | - Yu-Hsuan Tsai
- School of Chemistry, Cardiff University, Cardiff, Wales, United Kingdom
| |
Collapse
|
24
|
Serfling R, Seidel L, Bock A, Lohse MJ, Annibale P, Coin I. Quantitative Single-Residue Bioorthogonal Labeling of G Protein-Coupled Receptors in Live Cells. ACS Chem Biol 2019; 14:1141-1149. [PMID: 31074969 DOI: 10.1021/acschembio.8b01115] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
High-end microscopy studies of G protein-coupled receptors (GPCRs) require installing onto the receptors bright and photostable dyes. Labeling must occur in quantitative yields, to allow stoichiometric data analysis, and in a minimally invasive fashion, to avoid perturbing GPCR function. We demonstrate here that the genetic incorporation of trans-cyclooct-2-ene lysine (TCO*) allows achieving quantitative single-residue labeling of the extracellular loops of the β2-adrenergic and the muscarinic M2 class A GPCRs, as well as of the corticotropin releasing factor class B GPCR. Labeling occurs within a few minutes by reaction with dye-tetrazine conjugates on the surface of live cells and preserves the functionality of the receptors. To precisely quantify the labeling yields, we devise a method based on fluorescence fluctuation microscopy that extracts the number of labeling sites at the single-cell level. Further, we show that single-residue labeling is better suited for studies of GPCR diffusion than fluorescent-protein tags, since the latter can affect the mobility of the receptor. Finally, by performing dual-color competitive labeling on a single TCO* site, we devise a method to estimate the oligomerization state of a GPCR without the need for a biological monomeric reference, which facilitates the application of fluorescence methods to oligomerization studies. As TCO* and the dye-tetrazines used in this study are commercially available and the described microscopy techniques can be performed on a commercial microscope, we expect our approach to be widely applicable to fluorescence microscopy studies of membrane proteins in general.
Collapse
Affiliation(s)
- Robert Serfling
- University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Brüderstr. 34, 04103 Leipzig, Germany
| | - Lisa Seidel
- University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Brüderstr. 34, 04103 Leipzig, Germany
| | - Andreas Bock
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Martin J. Lohse
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Paolo Annibale
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Irene Coin
- University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Brüderstr. 34, 04103 Leipzig, Germany
| |
Collapse
|
25
|
Tong C, Liang H, Liu X, Yuan B, Xue B, Tong Z, Yin P. The protective effects of exenatide against AGEs-induced articular matrix degradation in human primary chondrocytes. Am J Transl Res 2019; 11:2081-2089. [PMID: 31105819 PMCID: PMC6511778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 01/04/2019] [Indexed: 06/09/2023]
Abstract
Osteoarthritis (OA) presents a major global health burden and is projected to become even more prevalent in coming decades. Therefore, it is of utmost importance to uncover novel therapies for the treatment and prevention of this disease. In the present study, we investigated the effects of exenatide, a specific glucagon-like peptide (GLP) agonist, on degradation of type II collagen and aggrecan, the two main components of the articular extracellular matrix, in human primary chondrocytes. Our results reveal that exenatide could ameliorate degradation of type II collagen and aggrecan by inhibiting expression of metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) induced by advanced glycation end-products. We also found that exenatide reduces oxidative stress and inhibits activation of nuclear factor-κB through the p38 cellular signaling pathway. Taken together, the findings of this study indicate that exenatide may have potential as a novel treatment for osteoarthritis.
Collapse
Affiliation(s)
- Changgui Tong
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Haidong Liang
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Xuehui Liu
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Bo Yuan
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Boqiong Xue
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Zhihong Tong
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| | - Peng Yin
- Department of Hand and Foot Microsurgery, The Second Hospital of Dalian Medical University Dalian 116023, Liaoning, China
| |
Collapse
|
26
|
Iacobucci C, Piotrowski C, Rehkamp A, Ihling CH, Sinz A. The First MS-Cleavable, Photo-Thiol-Reactive Cross-Linker for Protein Structural Studies. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:139-148. [PMID: 29679287 DOI: 10.1007/s13361-018-1952-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 06/08/2023]
Abstract
Cleavable cross-linkers are gaining increasing importance for chemical cross-linking/mass spectrometry (MS) as they permit a reliable and automated data analysis in structural studies of proteins and protein assemblies. Here, we introduce 1,3-diallylurea (DAU) as the first CID-MS/MS-cleavable, photo-thiol-reactive cross-linker. DAU is a commercially available, inexpensive reagent that efficiently undergoes an anti-Markovnikov hydrothiolation with cysteine residues in the presence of a radical initiator upon UV-A irradiation. Radical cysteine cross-linking proceeds via an orthogonal "click reaction" and yields stable alkyl sulfide products. DAU reacts at physiological pH and cross-linking reactions with peptides, and proteins can be performed at temperatures as low as 4 °C. The central urea bond is efficiently cleaved upon collisional activation during tandem MS experiments generating characteristic product ions. This improves the reliability of automated cross-link identification. Different radical initiators have been screened for the cross-linking reaction of DAU using the thiol-containing compounds cysteine and glutathione. Our concept has also been exemplified for the biologically relevant proteins bMunc13-2 and retinal guanylyl cyclase-activating protein-2. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Claudio Iacobucci
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle (Saale), Germany.
| | - Christine Piotrowski
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle (Saale), Germany
| | - Anne Rehkamp
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle (Saale), Germany
| | - Christian H Ihling
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle (Saale), Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120, Halle (Saale), Germany.
| |
Collapse
|
27
|
Glucagon-Like Peptide-1 Receptor Agonists and Cardiovascular Risk Reduction in Type 2 Diabetes Mellitus: Is It a Class Effect? Curr Cardiol Rep 2018; 20:113. [DOI: 10.1007/s11886-018-1051-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Slominsky PA, Shadrina MI. Peptide Pharmaceuticals: Opportunities, Prospects, and Limitations. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2018. [DOI: 10.3103/s0891416818010123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Huang Y, Liu T. Therapeutic applications of genetic code expansion. Synth Syst Biotechnol 2018; 3:150-158. [PMID: 30345400 PMCID: PMC6190509 DOI: 10.1016/j.synbio.2018.09.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 12/05/2022] Open
Abstract
In nature, a limited, conservative set of amino acids are utilized to synthesize proteins. Genetic code expansion technique reassigns codons and incorporates noncanonical amino acids (ncAAs) through orthogonal aminoacyl-tRNA synthetase (aaRS)/tRNA pairs. The past decade has witnessed the rapid growth in diversity and scope for therapeutic applications of this technology. Here, we provided an update on the recent progress using genetic code expansion in the following areas: antibody-drug conjugates (ADCs), bispecific antibodies (BsAb), immunotherapies, long-lasting protein therapeutics, biosynthesized peptides, engineered viruses and cells, as well as other therapeutic related applications, where the technique was used to elucidate the mechanisms of biotherapeutics and drug targets.
Collapse
Affiliation(s)
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
30
|
Simms J, Uddin R, Sakmar TP, Gingell JJ, Garelja ML, Hay DL, Brimble MA, Harris PW, Reynolds CA, Poyner DR. Photoaffinity Cross-Linking and Unnatural Amino Acid Mutagenesis Reveal Insights into Calcitonin Gene-Related Peptide Binding to the Calcitonin Receptor-like Receptor/Receptor Activity-Modifying Protein 1 (CLR/RAMP1) Complex. Biochemistry 2018; 57:4915-4922. [PMID: 30004692 DOI: 10.1021/acs.biochem.8b00502] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Calcitonin gene-related peptide (CGRP) binds to the complex of the calcitonin receptor-like receptor (CLR) with receptor activity-modifying protein 1 (RAMP1). How CGRP interacts with the transmembrane domain (including the extracellular loops) of this family B receptor remains unclear. In this study, a photoaffinity cross-linker, p-azido l-phenylalanine (azF), was incorporated into CLR, chiefly in the second extracellular loop (ECL2) using genetic code expansion and unnatural amino acid mutagenesis. The method was optimized to ensure efficient photolysis of azF residues near the transmembrane bundle of the receptor. A CGRP analogue modified with fluorescein at position 15 was used for detection of ultraviolet-induced cross-linking. The methodology was verified by confirming the known contacts of CGRP to the extracellular domain of CLR. Within ECL2, the chief contacts were I284 on the loop itself and L291, at the top of the fifth transmembrane helix (TM5). Minor contacts were noted along the lip of ECL2 between S286 and L290 and also with M223 in TM3 and F349 in TM6. Full length molecular models of the bound receptor complex suggest that CGRP sits at the top of the TM bundle, with Thr6 of the peptide making contacts with L291 and H295. I284 is likely to contact Leu12 and Ala13 of CGRP, and Leu16 of CGRP is at the ECL/extracellular domain boundary of CLR. The reduced potency, Emax, and affinity of [Leu16Ala]-human α CGRP are consistent with this model. Contacts between Thr6 of CGRP and H295 may be particularly important for receptor activation.
Collapse
Affiliation(s)
- John Simms
- Aston University , Birmingham B4 7ET , U.K
- Coventry University , Priory Street , Coventry CV1 5FB , U.K
| | | | - Thomas P Sakmar
- The Rockefeller University , 1230 York Avenue , New York , New York 10065 , United States
| | - Joseph J Gingell
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Michael L Garelja
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Debbie L Hay
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Margaret A Brimble
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | - Paul W Harris
- University of Auckland , 3A Symonds Street , Auckland 1010 , New Zealand
| | | | | |
Collapse
|
31
|
Coin I. Application of non-canonical crosslinking amino acids to study protein-protein interactions in live cells. Curr Opin Chem Biol 2018; 46:156-163. [PMID: 30077876 DOI: 10.1016/j.cbpa.2018.07.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/02/2018] [Accepted: 07/13/2018] [Indexed: 02/06/2023]
Abstract
The genetic incorporation of non-canonical amino acids (ncAAs) equipped with photo-crosslinking and chemical crosslinking moieties has found broad application in the study of protein-protein interactions from a unique perspective in live cells. We highlight here applications of photo-activatable ncAAs to map protein interaction surfaces and to capture protein-protein interactions, and we describe recent efforts to efficiently couple photo-crosslinking with mass spectrometric analysis. In addition, we describe recent advances in the development and application of ncAAs for chemical crosslinking, including protein stapling, photo-control of protein conformation, two-dimensional crosslinking, and stabilization of transient and low-affinity protein-protein interactions. We expect that the field will keep growing in the near future and enable the tackling of ambitious biological questions.
Collapse
Affiliation(s)
- Irene Coin
- University of Leipzig, Faculty of Life Sciences, Institute of Biochemistry, Brüderstr. 34, 04301 Leipzig, Germany.
| |
Collapse
|
32
|
Serfling R, Seidel L, Böttke T, Coin I. Optimizing the Genetic Incorporation of Chemical Probes into GPCRs for Photo-crosslinking Mapping and Bioorthogonal Chemistry in Live Mammalian Cells. J Vis Exp 2018. [PMID: 29683449 DOI: 10.3791/57069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The genetic incorporation of non-canonical amino acids (ncAAs) via amber stop codon suppression is a powerful technique to install artificial probes and reactive moieties onto proteins directly in the live cell. Each ncAA is incorporated by a dedicated orthogonal suppressor-tRNA/amino-acyl-tRNA-synthetase (AARS) pair that is imported into the host organism. The incorporation efficiency of different ncAAs can greatly differ, and be unsatisfactory in some cases. Orthogonal pairs can be improved by manipulating either the AARS or the tRNA. However, directed evolution of tRNA or AARS using large libraries and dead/alive selection methods are not feasible in mammalian cells. Here, a facile and robust fluorescence-based assay to evaluate the efficiency of orthogonal pairs in mammalian cells is presented. The assay allows screening tens to hundreds of AARS/tRNA variants with a moderate effort and within a reasonable time. Use of this assay to generate new tRNAs that significantly improve the efficiency of the pyrrolysine orthogonal system is described, along with the application of ncAAs to the study of G-protein coupled receptors (GPCRs), which are challenging objects for ncAA mutagenesis. First, by systematically incorporating a photo-crosslinking ncAA throughout the extracellular surface of a receptor, binding sites of different ligands on the intact receptor are mapped directly in the live cell. Second, by incorporating last-generation ncAAs into a GPCR, ultrafast catalyst-free receptor labeling with a fluorescent dye is demonstrated, which exploits bioorthogonal strain-promoted inverse Diels Alder cycloaddition (SPIEDAC) on the live cell. As ncAAs can be generally applied to any protein independently on its size, the method is of general interest for a number of applications. In addition, ncAA incorporation does not require any special equipment and is easily performed in standard biochemistry labs.
Collapse
Affiliation(s)
- Robert Serfling
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig
| | - Lisa Seidel
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig
| | - Thore Böttke
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig
| | - Irene Coin
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig;
| |
Collapse
|
33
|
Crnković A, Vargas-Rodriguez O, Merkuryev A, Söll D. Effects of Heterologous tRNA Modifications on the Production of Proteins Containing Noncanonical Amino Acids. Bioengineering (Basel) 2018; 5:bioengineering5010011. [PMID: 29393901 PMCID: PMC5874877 DOI: 10.3390/bioengineering5010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 12/21/2022] Open
Abstract
Synthesis of proteins with noncanonical amino acids (ncAAs) enables the creation of protein-based biomaterials with diverse new chemical properties that may be attractive for material science. Current methods for large-scale production of ncAA-containing proteins, frequently carried out in Escherichia coli, involve the use of orthogonal aminoacyl-tRNA synthetases (o-aaRSs) and tRNAs (o-tRNAs). Although o-tRNAs are designed to be orthogonal to endogenous aaRSs, their orthogonality to the components of the E. coli metabolism remains largely unexplored. We systematically investigated how the E. coli tRNA modification machinery affects the efficiency and orthogonality of o-tRNASep used for production of proteins with the ncAA O-phosphoserine (Sep). The incorporation of Sep into a green fluorescent protein (GFP) in 42 E. coli strains carrying deletions of single tRNA modification genes identified several genes that affect the o-tRNA activity. Deletion of cysteine desulfurase (iscS) increased the yield of Sep-containing GFP more than eightfold, while overexpression of dimethylallyltransferase MiaA and pseudouridine synthase TruB improved the specificity of Sep incorporation. These results highlight the importance of tRNA modifications for the biosynthesis of proteins containing ncAAs, and provide a novel framework for optimization of o-tRNAs.
Collapse
Affiliation(s)
- Ana Crnković
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | - Oscar Vargas-Rodriguez
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | - Anna Merkuryev
- Department of Chemistry, Yale University, New Haven, CT 06520, USA.
| | - Dieter Söll
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
- Department of Chemistry, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
34
|
Chen Y, Lu L, Ye S. Genetic Code Expansion and Optoproteomics. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:599-610. [PMID: 29259524 PMCID: PMC5733852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Nature has invented photoreceptor proteins that are involved in sensing and response to light in living organisms. Genetic code expansion (GCE) technology has provided new tools to transform light insensitive proteins into novel photoreceptor proteins. It is achieved by the site-specific incorporation of unnatural amino acids (Uaas) that carry light sensitive moieties serving as "pigments" that react to light via photo-decaging, cross-linking, or isomerization. Over the last two decades, various proteins including ion channels, GPCRs, transporters, and kinases have been successfully rendered light responsive owing to the functionalities of Uaas. Very recently, Cas9 protein has been engineered to enable light activation of genomic editing by CRISPR. Those novel proteins have not only led to discoveries of dynamic protein conformational changes with implications in diseases, but also facilitated the screening of ligand-protein and protein-protein interactions of pharmacological significance. This review covers the genetic editing principles for genetic code expansion and design concepts that guide the engineering of light-sensitive proteins. The applications have brought up a new concept of "optoproteomics" that, in contrast to "optogenetics," aims to combine optical methods and site-specific proteomics for investigating and intervening in biological functions.
Collapse
Affiliation(s)
- Yuting Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University (ECNU), Shanghai, China
| | - Linjie Lu
- Université Pierre-et-Marie-Curie, Laboratory of Computational and Quantitative Biology (LCQB), Institute of Biology Paris-Seine, Paris, France
| | - Shixin Ye
- Université Pierre-et-Marie-Curie, Laboratory of Computational and Quantitative Biology (LCQB), Institute of Biology Paris-Seine, Paris, France,Institut National de la Santé et de la Recherche Médicale, Paris, France,Centre National de la Recherche Scientifique, Paris, France,To whom all correspondence should be addressed: Shixin Ye-Lehmann, Laboratory of Computational and Quantitative Biology, Institute of Biology Paris-Seine, University of Pierre and Marie Curie, Bldg. C, 3rd floor, Room 311a, 4 Place Jussieu, 75006 Paris, France, Tel: 33.(0)1.44.27.60.57, .
| |
Collapse
|
35
|
Ramil CP, Dong M, An P, Lewandowski TM, Yu Z, Miller LJ, Lin Q. Spirohexene-Tetrazine Ligation Enables Bioorthogonal Labeling of Class B G Protein-Coupled Receptors in Live Cells. J Am Chem Soc 2017; 139:13376-13386. [PMID: 28876923 DOI: 10.1021/jacs.7b05674] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A new bioorthogonal reactant pair, spiro[2.3]hex-1-ene (Sph) and 3,6-di(2-pyridyl)-s-tetrazine (DpTz), for the strain-promoted inverse electron-demand Diels-Alder cycloaddition, that is, tetrazine ligation, is reported. As compared to the previously reported strained alkenes such as trans-cyclooctene (TCO) and 1,3-disubstituted cyclopropene, Sph exhibits balanced reactivity and stability in tetrazine ligation with the protein substrates. A lysine derivative of Sph, SphK, was site-selectively incorporated into the extracellular loop regions (ECLs) of GCGR and GLP-1R, two members of class B G protein-coupled receptors (GPCRs) in mammalian cells with the incorporation efficiency dependent on the location. Subsequent bioorthogonal reactions with the fluorophore-conjugated DpTz reagents afforded the fluorescently labeled GCGR and GLP-1R ECL mutants with labeling yield as high as 68%. A multitude of functional assays were performed with these GPCR mutants, including ligand binding, ligand-induced receptor internalization, and ligand-stimulated intracellular cAMP accumulation. Several positions in the ECL3s of GCGR and GLP-1R were identified that tolerate SphK mutagenesis and subsequent bioorthogonal labeling. The generation of functional, fluorescently labeled ECL3 mutants of GCGR and GLP-1R should allow biophysical studies of conformation dynamics of this important class of GPCRs in their native environment in live cells.
Collapse
Affiliation(s)
- Carlo P Ramil
- Department of Chemistry, State University of New York at Buffalo , Buffalo, New York 14260-3000, United States
| | - Maoqing Dong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic , Scottsdale, Arizona 85259, United States
| | - Peng An
- Department of Chemistry, State University of New York at Buffalo , Buffalo, New York 14260-3000, United States
| | - Tracey M Lewandowski
- Department of Chemistry, State University of New York at Buffalo , Buffalo, New York 14260-3000, United States
| | - Zhipeng Yu
- Department of Chemistry, State University of New York at Buffalo , Buffalo, New York 14260-3000, United States
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic , Scottsdale, Arizona 85259, United States
| | - Qing Lin
- Department of Chemistry, State University of New York at Buffalo , Buffalo, New York 14260-3000, United States
| |
Collapse
|