1
|
Achouba Y, Peres B, Ascoët S, Meudal H, Caumes C, Zoukimian C, Millet H, Choteau-Bodor M, Carvalhosa C, Croyal M, Bouchama F, Wulff H, Téletchéa S, Béroud R, Ishow E, Landon C, Boumendjel A, Montnach J, De Waard M. Photoisomerization of Azobenzene-Extended Charybdotoxin for the Optical Control of K v1.2 Potassium Channel Activity. Angew Chem Int Ed Engl 2025; 64:e202423278. [PMID: 40013552 DOI: 10.1002/anie.202423278] [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: 11/28/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 02/28/2025]
Abstract
Natural peptides from animal venoms effectively modulate ion channel activity. While photoswitches regulate small compound pharmacology, their application to natural peptides rich in disulfide bridges and active on ion channels is novel due to larger pharmacophores. A pilot study integrating azobenzene photoswitches into charybdotoxin (ChTx), known for blocking potassium channels is initiated. Two click-chemistry-compatible azobenzene are synthesized differing in length and amide orientation (Az1 & Az2). Az1 is grafted onto ChTx at various amino acid positions using L-azidohomoalanine mutation. ChTx monomers outperformed dimers, particularly with azobenzene at position 14, by exhibiting optimal photoswitching activity. In the cis configuration, Az1 altered ChTx's pharmacophore, reducing potassium channel blockage, while conversely, Az2 increased ChTx potency. This study pioneers photoswitch application to complex peptides, leveraging structure-activity relationships. Successful integration depends on precise azobenzene positioning and chemical grafting guided by SAR insights. This advancement underscores the adaptability of photoswitch technology to intricate peptide structures, offering new avenues for pharmacological modulation.
Collapse
Affiliation(s)
- Yanis Achouba
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | - Basile Peres
- Département de Pharmacochimie Moléculaire, Université Grenoble Alpes, CNRS, Grenoble, F-38000, France
| | - Steven Ascoët
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | - Hervé Meudal
- Center for Molecular Biophysics, CNRS, Orléans, 45071, France
| | | | | | - Hugo Millet
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | | | | | - Mikael Croyal
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | - Fella Bouchama
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, CA, USA
| | | | - Rémy Béroud
- Smartox Biotechnology, Saint-Egrève, 38120, France
- SB-Peptides, Saint-Egrève, 38120, France
| | - Eléna Ishow
- CEISAM, Nantes Université, CNRS, Nantes, 44322, France
| | - Céline Landon
- Center for Molecular Biophysics, CNRS, Orléans, 45071, France
| | | | - Jérôme Montnach
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
| | - Michel De Waard
- L'institut du thorax, Nantes Université, CNRS, INSERM, Nantes, F-44000, France
- Smartox Biotechnology, Saint-Egrève, 38120, France
- Laboratory of Excellence «Ion Channels, Science and Therapeutics», Valbonne, F-06560, France
| |
Collapse
|
2
|
El-Gaby MSA, Abdel Reheim MAM, Akrim ZSM, Naguib BH, Saleh NM, El-Adasy ABAAM, El-Adl K, Mohamady S. 2-Thioxo-3,4-dihydropyrimidine and thiourea endowed with sulfonamide moieties as dual EGFR T790M and VEGFR-2 inhibitors: Design, synthesis, docking, and anticancer evaluations. Drug Dev Res 2024; 85:e22143. [PMID: 38349267 DOI: 10.1002/ddr.22143] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/16/2023] [Indexed: 02/15/2024]
Abstract
The effectiveness of a new series of thiopyrimidine and thiourea containing sulfonamides moieties was tested on HCT-116, MCF-7, HepG2, and A549. HepG2 cell line was the one that all the new derivatives affected the most. The greatest potent compounds against the four HepG2, HCT116, MCF-7, and A549 cell lines were 8f and 8g with IC50 = 4.13, 6.64, 5.74, 6.85 µM and 4.09, 4.36, 4.22, 7.25 µM correspondingly. Compound 8g exhibited higher activity than sorafenib against HCT116 and MCF-7 but exhibited lower activity against HepG2 and A549. Moreover, compounds 8f and 8g exhibited higher activities than erlotinib on HepG2, HCT116, and MCF-7 but demonstrated lower activity on A549. The most potent cytotoxic derivatives 6f, 6g, 8c, 8d, 8e, 8f, and 8g were examined on normal VERO cell lines. Our derivatives have low toxicity on VERO cells with IC50 values ranging from 32.05 to 53.15 μM. Additionally, all compounds were assessed for dual VEGFR-2 and EGFRT790M inhibition effects. Compounds 8f and 8g were the most potent derivatives inhibited VEGFR-2 at IC50 value of 0.88 and 0.90 µM, correspondingly. As well, derivatives 8f and 8g could inhibit EGFRT790M demonstrating strongest effects with IC50 = 0.32 and 0.33 µM sequentially. Additionally, the greatest active derivatives ADMET profile was evaluated in relationship with sorafenib and erlotinib as reference agents. The data attained from docking were greatly related to that achieved from the biological testing.
Collapse
Affiliation(s)
- Mohamed S A El-Gaby
- Department of Chemistry, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| | | | - Zuhir S M Akrim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Omar Almukhtar University Al-Bayda, Libya
| | - Bassem H Naguib
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Nashwa M Saleh
- Department of Chemistry, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | | | - Khaled El-Adl
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Samy Mohamady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
3
|
Anwer KE, El-Hddad SSA, Abd El-Sattar NEA, El-Morsy A, Khedr F, Mohamady S, Keshek DE, Salama SA, El-Adl K, Hanafy NS. Five and six membered heterocyclic rings endowed with azobenzene as dual EGFR T790M and VEGFR-2 inhibitors: design, synthesis, in silico ADMET profile, molecular docking, dynamic simulation and anticancer evaluations. RSC Adv 2023; 13:35321-35338. [PMID: 38053688 PMCID: PMC10695193 DOI: 10.1039/d3ra06614b] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023] Open
Abstract
Novel azobenzene scaffold-joined heterocyclic isoxazole, pyrazole, triazole, and/or triazine moieties have been developed and synthesized utilizing microwave and traditional methods. Our compounds were tested for growth inhibition of A549, MCF-7, HCT-116, and HepG2 tumors by dual targeting the VEGFR-2 and EGFRT790M enzymes. The suggested compound's manner of binding with EGFRT790M and VEGFR-2 active sites was explored through molecular design and MD modeling. The information from the results of the biological screening and the docking studies was highly correlated. The A549 cell line was the one that responded to the novel compound's effects most effectively. Having IC50 values of 5.15, 6.37, 8.44 and 6.23 μM, respectively, 14 was the most effective derivative on the four A549, MCF-7, HCT116 and HepG2 cancer cells. It had greater activity than erlotinib and slightly inferior activities on the tested cell lines than sorafenib, respectively. The cytotoxicity of the most effective derivatives, 5, 6, 10 and 14, was evaluated against typical VERO cell lines. Having IC50 values ranging from 42.32 to 55.20 μM, the results showed that the investigated drugs have modest toxicity against VERO normal cells. Additionally all derivatives were assessed for their dual VEGFR-2 and EGFRT790M inhibitory effects. Among them, derivatives 14, 5 and 10 were established as the greatest inhibitors of VEGFR-2 at IC50 values of 0.95, 1.25 and 1.50 μM correspondingly. As well, derivatives 14, 6, 5 and 10 could inhibit EGFRT790M activity demonstrating strongest effects with IC50 = 0.25, 0.35, 0.40 and 0.50 μM respectively. Furthermore, the ADMET profile was evaluated for compounds 5, 6, 10 and 14 in contrast to reference drugs sorafenib and erlotinib.
Collapse
Affiliation(s)
- Kurls E Anwer
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
| | | | - Nour E A Abd El-Sattar
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
- Basic & Medical Sciences Department, Faculty of Dentistry, Alryada University for Science & Technology Egypt
| | - Ahmed El-Morsy
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University Najaf Iraq
| | - Fathalla Khedr
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Samy Mohamady
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt Cairo Egypt
| | - Doaa E Keshek
- Department of Biology, Jumum College University, Umm Al-Qura University P.O. Box 7388 Makkah 21955 Sudia Arabia
- Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Centre Giza Egypt
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Kingdom of Saudi Arabia
| | - Khaled El-Adl
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University Nasr City 11884 Cairo Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
| | - Noura S Hanafy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
| |
Collapse
|
4
|
Simon I, Homan EJ, Wijtmans M, Sundström M, Leurs R, De Esch IJP, Zarzycka BA. PSW-Designer: An Open-Source Computational Platform for the Design and Virtual Screening of Photopharmacological Ligands. J Chem Inf Model 2023; 63:6696-6705. [PMID: 37831965 PMCID: PMC10647043 DOI: 10.1021/acs.jcim.3c01050] [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: 07/11/2023] [Indexed: 10/15/2023]
Abstract
Photoswitchable (PSW) molecules offer an attractive opportunity for the optical control of biological processes. However, the successful design of such compounds remains a challenging multioptimization endeavor, resulting in several biological target classes still relatively poorly explored by photoswitchable ligands, as is the case for G protein-coupled receptors (GPCRs). Here, we present the PSW-Designer, a fully open-source computational platform, implemented in the KNIME Analytics Platform, to design and virtually screen novel photoswitchable ligands for photopharmacological applications based on privileged scaffolds. We demonstrate the applicability of the PSW-Designer to GPCRs and assess its predictive capabilities via two retrospective case studies. Furthermore, by leveraging bioactivity information on known ligands, typical and atypical strategies for photoswitchable group incorporation, and the increasingly structural information available for biological targets, the PSW-Design will facilitate the design of novel photoswitchable molecules with improved photopharmacological properties and increased binding affinity shifts upon illumination for GPCRs and many other protein targets.
Collapse
Affiliation(s)
- Icaro
A. Simon
- Division
of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for
Molecular and Life Sciences, Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Evert J. Homan
- Science
for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Maikel Wijtmans
- Division
of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for
Molecular and Life Sciences, Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Michael Sundström
- Centre
for Molecular Medicine, Karolinska Institutet, S-171 76 Stockholm, Sweden
| | - Rob Leurs
- Division
of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for
Molecular and Life Sciences, Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J. P. De Esch
- Division
of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for
Molecular and Life Sciences, Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Barbara A. Zarzycka
- Division
of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for
Molecular and Life Sciences, Vrije Universiteit
Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
5
|
Marcus DJ, Bruchas MR. Optical Approaches for Investigating Neuromodulation and G Protein-Coupled Receptor Signaling. Pharmacol Rev 2023; 75:1119-1139. [PMID: 37429736 PMCID: PMC10595021 DOI: 10.1124/pharmrev.122.000584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 07/12/2023] Open
Abstract
Despite the fact that roughly 40% of all US Food and Drug Administration (FDA)-approved pharmacological therapeutics target G protein-coupled receptors (GPCRs), there remains a gap in our understanding of the physiologic and functional role of these receptors at the systems level. Although heterologous expression systems and in vitro assays have revealed a tremendous amount about GPCR signaling cascades, how these cascades interact across cell types, tissues, and organ systems remains obscure. Classic behavioral pharmacology experiments lack both the temporal and spatial resolution to resolve these long-standing issues. Over the past half century, there has been a concerted effort toward the development of optical tools for understanding GPCR signaling. From initial ligand uncaging approaches to more recent development of optogenetic techniques, these strategies have allowed researchers to probe longstanding questions in GPCR pharmacology both in vivo and in vitro. These tools have been employed across biologic systems and have allowed for interrogation of everything from specific intramolecular events to pharmacology at the systems level in a spatiotemporally specific manner. In this review, we present a historical perspective on the motivation behind and development of a variety of optical toolkits that have been generated to probe GPCR signaling. Here we highlight how these tools have been used in vivo to uncover the functional role of distinct populations of GPCRs and their signaling cascades at a systems level. SIGNIFICANCE STATEMENT: G protein-coupled receptors (GPCRs) remain one of the most targeted classes of proteins for pharmaceutical intervention, yet we still have a limited understanding of how their unique signaling cascades effect physiology and behavior at the systems level. In this review, we discuss a vast array of optical techniques that have been devised to probe GPCR signaling both in vitro and in vivo.
Collapse
Affiliation(s)
- David J Marcus
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| |
Collapse
|
6
|
Tian H, You S, Xiong T, Ji M, Zhang K, Jiang L, Du T, Li Y, Liu W, Lin S, Chen X, Xu H. Discovery of a Novel Photocaged PI3K Inhibitor Capable of Real-Time Reporting of Drug Release. ACS Med Chem Lett 2023; 14:1100-1107. [PMID: 37583818 PMCID: PMC10424311 DOI: 10.1021/acsmedchemlett.3c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
A novel photocaged PI3K inhibitor 2 was designed and synthesized by introducing a cascade photocaging group to block its key interaction with the kinase. Upon UV light irradiation, the photocaged compound released a highly potent PI3K inhibitor to recover its anticancer properties and a fluorescent dye for real-time reporting of drug release, providing a new approach for studying the PI3K signaling transduction pathway as well as developing precisely controlled cancer therapeutics.
Collapse
Affiliation(s)
- Hua Tian
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Shen You
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Tianning Xiong
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Ming Ji
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Kehui Zhang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Lin Jiang
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Tingting Du
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Ying Li
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Wenqian Liu
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Songwen Lin
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Xiaoguang Chen
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| | - Heng Xu
- State
Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Beijing
Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing 100050, China
- Key
Laboratory of Small Molecule Immuno-Oncology Drug Discovery, Chinese Academy of Medical Sciences and Peking Union
Medical College, Beijing 100050, China
| |
Collapse
|
7
|
Pereira V, Arias JA, Llebaria A, Goudet C. Photopharmacological manipulation of amygdala metabotropic glutamate receptor mGlu4 alleviates neuropathic pain. Pharmacol Res 2023; 187:106602. [PMID: 36529205 DOI: 10.1016/j.phrs.2022.106602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Neuropathic pain is a common health problem resulting in exacerbated response to noxious and non noxious stimuli, as well as impaired emotional and cognitive responses. Unfortunately, neuropathic pain is also one of the most difficult pain syndromes to manage, highlighting the importance of better understanding the brain regions and neuromodulatory mechanisms involved in its regulation. Among the many interconnected brain areas which process pain, the amygdala is known to play an important role in the integration of sensory and emotional pain signals. Here we questioned the ability of a recently identified neuromodulatory mechanism associated with the metabotropic glutamate receptors mGlu4 in the amygdala to modulate neuropathic pain. In a murine model of peripheral mononeuropathy, we demonstrate that pharmacological activation of amygdala mGlu4 efficiently alleviates sensory and depressive-like symptoms in both male and female mice. Moreover, we reveal a differential modulation of these symptoms. Activating mGlu4 in the contralateral amygdala relative to the side of the mononeuropathy, is necessary and sufficient to relieve both sensory and depressive-like symptoms, while ipsilateral activation solely reduces depressive-like symptoms. Furthermore, using photopharmacology, a recent strategy allowing precise photocontrol of endogenous proteins, we further demonstrate the dynamic alleviation of neuropathic pain through light-dependent facilitation of mGlu4 by a photoswitchable positive allosteric modulator. Finally, coupling photopharmacology and analgesic conditioned place preference, we show a significant pain-reducing effect of mGlu4 activation. Taken together, these data highlight the analgesic potential of enhancing amygdala mGlu4 activity to counteract neuropathy reinforcing its therapeutic interest for the treatment of pathological pain.
Collapse
Affiliation(s)
| | | | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Cyril Goudet
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
8
|
Albanese P, Cataldini S, Ren CZJ, Valletti N, Brunetti J, Chen JLY, Rossi F. Light-Switchable Membrane Permeability in Giant Unilamellar Vesicles. Pharmaceutics 2022; 14:2777. [PMID: 36559270 PMCID: PMC9780837 DOI: 10.3390/pharmaceutics14122777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
In this work, giant unilamellar vesicles (GUVs) were synthesized by blending the natural phospholipid 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) with a photoswitchable amphiphile (1) that undergoes photoisomerization upon irradiation with UV-A (E to Z) and blue (Z to E) light. The mixed vesicles showed marked changes in behavior in response to UV light, including changes in morphology and the opening of pores. The fine control of membrane permeability with consequent cargo release could be attained by modulating either the UV irradiation intensity or the membrane composition. As a proof of concept, the photocontrolled release of sucrose from mixed GUVs is demonstrated using microscopy (phase contrast) and confocal studies. The permeability of the GUVs to sucrose could be increased to ~4 × 10-2 μm/s when the system was illuminated by UV light. With respect to previously reported systems (entirely composed of synthetic amphiphiles), our findings demonstrate the potential of photosensitive GUVs that are mainly composed of natural lipids to be used in medical and biomedical applications, such as targeted drug delivery and localized topical treatments.
Collapse
Affiliation(s)
- Paola Albanese
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| | - Simone Cataldini
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, Via Aldo Moro, 53100 Siena, Italy
| | - Chloe Z-J Ren
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology, Auckland 1142, New Zealand
| | - Nadia Valletti
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| | - Jlenia Brunetti
- MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Jack L-Y Chen
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, Via Aldo Moro, 53100 Siena, Italy
- Centre for Biomedical and Chemical Sciences, School of Science, Auckland University of Technology, Auckland 1142, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Federico Rossi
- Department of Earth, Environmental & Physical Sciences, University of Siena, Pian Mantellini 44, 53100 Siena, Italy
| |
Collapse
|
9
|
Matera C, Calvé P, Casadó-Anguera V, Sortino R, Gomila AMJ, Moreno E, Gener T, Delgado-Sallent C, Nebot P, Costazza D, Conde-Berriozabal S, Masana M, Hernando J, Casadó V, Puig MV, Gorostiza P. Reversible Photocontrol of Dopaminergic Transmission in Wild-Type Animals. Int J Mol Sci 2022; 23:ijms231710114. [PMID: 36077512 PMCID: PMC9456102 DOI: 10.3390/ijms231710114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 01/09/2023] Open
Abstract
Understanding the dopaminergic system is a priority in neurobiology and neuropharmacology. Dopamine receptors are involved in the modulation of fundamental physiological functions, and dysregulation of dopaminergic transmission is associated with major neurological disorders. However, the available tools to dissect the endogenous dopaminergic circuits have limited specificity, reversibility, resolution, or require genetic manipulation. Here, we introduce azodopa, a novel photoswitchable ligand that enables reversible spatiotemporal control of dopaminergic transmission. We demonstrate that azodopa activates D1-like receptors in vitro in a light-dependent manner. Moreover, it enables reversibly photocontrolling zebrafish motility on a timescale of seconds and allows separating the retinal component of dopaminergic neurotransmission. Azodopa increases the overall neural activity in the cortex of anesthetized mice and displays illumination-dependent activity in individual cells. Azodopa is the first photoswitchable dopamine agonist with demonstrated efficacy in wild-type animals and opens the way to remotely controlling dopaminergic neurotransmission for fundamental and therapeutic purposes.
Collapse
Affiliation(s)
- Carlo Matera
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Pablo Calvé
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Rosalba Sortino
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Alexandre M. J. Gomila
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Thomas Gener
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Cristina Delgado-Sallent
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Pau Nebot
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Davide Costazza
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
| | - Sara Conde-Berriozabal
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERNED, 08036 Barcelona, Spain
| | - Mercè Masana
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Institute of Neuroscience, University of Barcelona, IDIBAPS, CIBERNED, 08036 Barcelona, Spain
| | - Jordi Hernando
- Department of Chemistry, Autonomous University of Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - M. Victoria Puig
- Hospital del Mar Medical Research Institute (IMIM), Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute for Science and Technology, 08028 Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
10
|
Somalo-Barranco G, Serra C, Lyons D, Piggins HD, Jockers R, Llebaria A. Design and Validation of the First Family of Photo-Activatable Ligands for Melatonin Receptors. J Med Chem 2022; 65:11229-11240. [PMID: 35930058 PMCID: PMC9421648 DOI: 10.1021/acs.jmedchem.2c00717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
![]()
Melatonin is a neurohormone released in a circadian manner
with
peak levels at night. Melatonin mediates its effects mainly through
G protein-coupled MT1 and MT2 receptors. Drugs
acting on melatonin receptors are indicated for circadian rhythm-
and sleep-related disorders. Tools to study the activation of these
receptors with high temporal resolution are lacking. Here, we synthesized
a family of light-activatable caged compounds by attaching o-nitrobenzyl (o-NB) or coumarin photocleavable
groups to melatonin indolic nitrogen. All caged compounds showed the
expected decrease in binding affinity for MT1 and MT2. The o-NB derivative MCS-0382 showed the
best uncaging and biological properties, with 250-fold increase in
affinity and potency upon illumination. Generation of melatonin from
MCS-0382 was further demonstrated by its ability to modulate the excitation
of SCN neurons in rat brain slices. MCS-0382 is available to study
melatonin effects in a temporally controlled manner in cellular and
physiological settings.
Collapse
Affiliation(s)
- Gloria Somalo-Barranco
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France.,MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Carme Serra
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain.,SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - David Lyons
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BS8 1TD Bristol, U.K
| | - Hugh D Piggins
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BS8 1TD Bristol, U.K
| | - Ralf Jockers
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| |
Collapse
|
11
|
Duran‐Corbera A, Faria M, Ma Y, Prats E, Dias A, Catena J, Martinez KL, Raldua D, Llebaria A, Rovira X. A Photoswitchable Ligand Targeting the β
1
‐Adrenoceptor Enables Light‐Control of the Cardiac Rhythm**. Angew Chem Int Ed Engl 2022; 61:e202203449. [PMID: 35608051 PMCID: PMC9401038 DOI: 10.1002/anie.202203449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Indexed: 11/06/2022]
Abstract
Catecholamine‐triggered β‐adrenoceptor (β‐AR) signaling is essential for the correct functioning of the heart. Although both β1‐ and β2‐AR subtypes are expressed in cardiomyocytes, drugs selectively targeting β1‐AR have proven this receptor as the main target for the therapeutic effects of beta blockers in the heart. Here, we report a new strategy for the light‐control of β1‐AR activation by means of photoswitchable drugs with a high level of β1‐/β2‐AR selectivity. All reported molecules allow for an efficient real‐time optical control of receptor function in vitro. Moreover, using confocal microscopy we demonstrate that the binding of our best hit, pAzo‐2, can be reversibly photocontrolled. Strikingly, pAzo‐2 also enables a dynamic cardiac rhythm management on living zebrafish larvae using light, thus highlighting the therapeutic and research potential of the developed photoswitches. Overall, this work provides the first proof of precise control of the therapeutic target β1‐AR in native environments using light.
Collapse
Affiliation(s)
- Anna Duran‐Corbera
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Melissa Faria
- Institute for Environmental Assessment and Water Research (IDAEA), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Yuanyuan Ma
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Eva Prats
- Research and Development Center (CID), CSIC Jordi Girona 18 08034 Barcelona Spain
| | - André Dias
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Juanlo Catena
- SIMchem, Service of Synthesis of High Added Value Molecules Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 Barcelona Spain
| | - Karen L. Martinez
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Demetrio Raldua
- Institute for Environmental Assessment and Water Research (IDAEA), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Xavier Rovira
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
- Previous address: Molecular Photopharmacology Research Group The Tissue Repair and Regeneration Laboratory (TR2Lab) Faculty of Sciences and Technology University of Vic, Central University of Catalonia 08500 Vic Spain
| |
Collapse
|
12
|
Duran‐Corbera A, Faria M, Ma Y, Prats E, Dias A, Catena J, Martinez KL, Raldua D, Llebaria A, Rovira X. A Photoswitchable Ligand Targeting the β
1
‐Adrenoceptor Enables Light‐Control of the Cardiac Rhythm**. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anna Duran‐Corbera
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Melissa Faria
- Institute for Environmental Assessment and Water Research (IDAEA), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Yuanyuan Ma
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Eva Prats
- Research and Development Center (CID), CSIC Jordi Girona 18 08034 Barcelona Spain
| | - André Dias
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Juanlo Catena
- SIMchem, Service of Synthesis of High Added Value Molecules Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 Barcelona Spain
| | - Karen L. Martinez
- Department of Chemistry & Nanoscience Center University of Copenhagen Thorvaldsensvej 40 1871 Frederiksberg Denmark
| | - Demetrio Raldua
- Institute for Environmental Assessment and Water Research (IDAEA), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
| | - Xavier Rovira
- MCS, Laboratory of Medicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC), CSIC Jordi Girona, 18 08034 Barcelona Spain
- Molecular Photopharmacology Research Group The Tissue Repair and Regeneration Laboratory (TR2Lab) Faculty of Sciences and Technology University of Vic, Central University of Catalonia 08500 Vic Spain
| |
Collapse
|
13
|
Optical control of Class A G protein-coupled receptors with photoswitchable ligands. Curr Opin Pharmacol 2022; 63:102192. [DOI: 10.1016/j.coph.2022.102192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/26/2022]
|
14
|
Zhu J, Guo T, Wang Z, Zhao Y. Triggered azobenzene-based prodrugs and drug delivery systems. J Control Release 2022; 345:475-493. [PMID: 35339578 DOI: 10.1016/j.jconrel.2022.03.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 12/18/2022]
Abstract
Azobenzene-based molecules show unique trans-cis isomerization upon ultraviolet light irradiation, which induce the change of polarity, crystallinity, stability, and binding affinity with pharmacological target. Moreover, azobenzene is the substrate of azoreductase that is often overexpressed in many pathological sites, e.g. hypoxic solid tumor. Therefore, azobenzene can be a multifunctional molecule in material science, pharmaceutical science and biomedicine because of its sensitivity to light, hypoxia and certain enzymes, hence showing potential application in site-specific smart therapy. Herein we focus on the employment of azobenzene and its derivatives for engineering triggered prodrug and drug delivery systems, and provide an overview of photoswitchable azo-based prodrugs, the associated problems regarding ultraviolet light and reversible isomerization, as well as the potential solutions. We also present the advance of azo-bearing delivery vehicles wherein azobenzene act as the linker, capping agent, and building block, and discuss the corresponding mechanisms for controlled cargo release, endocytosis enhancement and sensitization of free radical cancer therapy.
Collapse
Affiliation(s)
- Jundong Zhu
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
15
|
Agonists and allosteric modulators promote signaling from different metabotropic glutamate receptor 5 conformations. Cell Rep 2021; 36:109648. [PMID: 34469715 PMCID: PMC8424648 DOI: 10.1016/j.celrep.2021.109648] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/08/2021] [Accepted: 08/11/2021] [Indexed: 11/20/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are dimeric G-protein-coupled receptors activated by the main excitatory neurotransmitter, L-glutamate. mGluR activation by agonists binding in the venus flytrap domain is regulated by positive (PAM) or negative (NAM) allosteric modulators binding to the 7-transmembrane domain (7TM). We report the cryo-electron microscopy structures of fully inactive and intermediate-active conformations of mGlu5 receptor bound to an antagonist and a NAM or an agonist and a PAM, respectively, as well as the crystal structure of the 7TM bound to a photoswitchable NAM. The agonist induces a large movement between the subunits, bringing the 7TMs together and stabilizing a 7TM conformation structurally similar to the inactive state. Using functional approaches, we demonstrate that the PAM stabilizes a 7TM active conformation independent of the conformational changes induced by agonists, representing an alternative mode of mGlu activation. These findings provide a structural basis for different mGluR activation modes. Cryo-EM analysis of thermostabilized mGlu5 receptor bound to inhibitors or activators X-ray structure of trans-Alloswitch-1 bound to thermostable mGlu5 7TMs Photopharmacology provides insight into allosteric regulation of mGlu5 7TMs Multiple conformations of mGlu5 receptor activate G protein
Collapse
|
16
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2021; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
17
|
Ricart-Ortega M, Berizzi AE, Pereira V, Malhaire F, Catena J, Font J, Gómez-Santacana X, Muñoz L, Zussy C, Serra C, Rovira X, Goudet C, Llebaria A. Mechanistic Insights into Light-Driven Allosteric Control of GPCR Biological Activity. ACS Pharmacol Transl Sci 2020; 3:883-895. [PMID: 33073188 DOI: 10.1021/acsptsci.0c00054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 12/31/2022]
Abstract
G protein-coupled receptors (GPCR), including the metabotrobic glutamate 5 receptor (mGlu5), are important therapeutic targets and the development of allosteric ligands for targeting GPCRs has become a desirable approach toward modulating receptor activity. Traditional pharmacological approaches toward modulating GPCR activity are still limited since precise spatiotemporal control of a ligand is lost as soon as it is administered. Photopharmacology proposes the use of photoswitchable ligands to overcome this limitation, since their activity can be reversibly controlled by light with high precision. As this is still a growing field, our understanding of the molecular mechanisms underlying the light-induced changes of different photoswitchable ligand pharmacology is suboptimal. For this reason, we have studied the mechanisms of action of alloswitch-1 and MCS0331; two freely diffusible, mGlu5 phenylazopyridine photoswitchable negative allosteric modulators. We combined photochemical, cell-based, and in vivo photopharmacological approaches to investigate the effects of trans-cis azobenzene photoisomerization on the functional activity and binding ability of these ligands to the mGlu5 allosteric pocket. From these results, we conclude that photoisomerization can take place inside and outside the ligand binding pocket, and this leads to a reversible loss in affinity, in part, due to changes in dissociation rates from the receptor. Ligand activity for both photoswitchable ligands deviates from high-affinity mGlu5 negative allosteric modulation (in the trans configuration) to reduced affinity for the mGlu5 in their cis configuration. Importantly, this mechanism translates to dynamic and reversible control over pain following local injection and illumination of negative allosteric modulators into a brain region implicated in pain control.
Collapse
Affiliation(s)
- Maria Ricart-Ortega
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain.,IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Alice E Berizzi
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Vanessa Pereira
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Fanny Malhaire
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Juanlo Catena
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Joan Font
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | | | - Lourdes Muñoz
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain.,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Charleine Zussy
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Carmen Serra
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain.,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Xavier Rovira
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| | - Cyril Goudet
- IGF, CNRS, INSERM, University of Montpellier, F-34094 Montpellier, France
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain.,SIMchem, Service of Synthesis of High Added Value Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona 08034, Spain
| |
Collapse
|
18
|
Shchepinova MM, Hanyaloglu AC, Frost GS, Tate EW. Chemical biology of noncanonical G protein-coupled receptor signaling: Toward advanced therapeutics. Curr Opin Chem Biol 2020; 56:98-110. [PMID: 32446179 DOI: 10.1016/j.cbpa.2020.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling membrane proteins, are the target of more than 30% of the drugs on the market. Recently, it has become clear that GPCR functions are far more multidimensional than previously thought, with multiple noncanonical aspects coming to light, including biased, oligomeric, and compartmentalized signaling. These additional layers of functional selectivity greatly expand opportunities for advanced therapeutic interventions, but the development of new chemical biology tools is absolutely required to improve our understanding of noncanonical GPCR regulation and pave the way for future drugs. In this opinion, we highlight the most notable examples of chemical and chemogenetic tools addressing new paradigms in GPCR signaling, discuss their promises and limitations, and explore future directions.
Collapse
Affiliation(s)
- Maria M Shchepinova
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Dept. Surgery and Cancer, Imperial College, London, UK
| | - Gary S Frost
- Department of Medicine, Faculty of Medicine, Nutrition and Dietetic Research Group, Imperial College, London, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, 80 Wood Lane, London, W12 0BZ, UK.
| |
Collapse
|