1
|
Tabanelli R, Brogi S, Calderone V. Targeting Opioid Receptors in Addiction and Drug Withdrawal: Where Are We Going? Int J Mol Sci 2023; 24:10888. [PMID: 37446064 PMCID: PMC10341731 DOI: 10.3390/ijms241310888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
This review article offers an outlook on the use of opioids as therapeutics for treating several diseases, including cancer and non-cancer pain, and focuses the analysis on the opportunity to target opioid receptors for treating opioid use disorder (OUD), drug withdrawal, and addiction. Unfortunately, as has been well established, the use of opioids presents a plethora of side effects, such as tolerance and physical and physiological dependence. Accordingly, considering the great pharmacological potential in targeting opioid receptors, the identification of opioid receptor ligands devoid of most of the adverse effects exhibited by current therapeutic agents is highly necessary. To this end, herein, we analyze some interesting molecules that could potentially be useful for treating OUD, with an in-depth analysis regarding in vivo studies and clinical trials.
Collapse
Affiliation(s)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (V.C.)
| | | |
Collapse
|
2
|
Intranuovo F, Brunetti L, DelRe P, Mangiatordi GF, Stefanachi A, Laghezza A, Niso M, Leonetti F, Loiodice F, Ligresti A, Kostrzewa M, Brea J, Loza MI, Sotelo E, Saviano M, Colabufo NA, Riganti C, Abate C, Contino M. Development of N-(1-Adamantyl)benzamides as Novel Anti-Inflammatory Multitarget Agents Acting as Dual Modulators of the Cannabinoid CB2 Receptor and Fatty Acid Amide Hydrolase. J Med Chem 2023; 66:235-250. [PMID: 36542836 DOI: 10.1021/acs.jmedchem.2c01084] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cannabinoid type 2 receptor (CB2R), belonging to the endocannabinoid system, is overexpressed in pathologies characterized by inflammation, and its activation counteracts inflammatory states. Fatty acid amide hydrolase (FAAH) is an enzyme responsible for the degradation of the main endocannabinoid anandamide; thus, the simultaneous CB2R activation and FAAH inhibition may be a synergistic anti-inflammatory strategy. Encouraged by principal component analysis (PCA) data identifying a wide chemical space shared by CB2R and FAAH ligands, we designed a small library of adamantyl-benzamides, as potential dual agents, CB2R agonists, and FAAH inhibitors. The new compounds were tested for their CB2R affinity/selectivity and CB2R and FAAH activity. Derivatives 13, 26, and 27, displaying the best pharmacodynamic profile as CB2R full agonists and FAAH inhibitors, decreased pro-inflammatory and increased anti-inflammatory cytokines production. Molecular docking simulations complemented the experimental findings by providing a molecular rationale behind the observed activities. These multitarget ligands constitute promising anti-inflammatory agents.
Collapse
Affiliation(s)
- Francesca Intranuovo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Leonardo Brunetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Pietro DelRe
- Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | | | - Angela Stefanachi
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Via Campi Flegrei 34, Pozzuoli 80078, Italy
| | - Jose Brea
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Maria Isabel Loza
- Innopharma Screening Platform, BioFarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain.,Department of Pharmacology, Pharmacy and Pharmaceutical Technology. School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Eddy Sotelo
- ComBioMed Research Group, Centro de Química Biológica y Materiales Moleculares (CIQUS), University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Michele Saviano
- Institute of Crystallography, National Research Council of Italy, Via Vivaldi, 43, Caserta 81100, Italy
| | - Nicola Antonio Colabufo
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Chiara Riganti
- Dipartimento di Oncologia, Università Degli Studi di Torino, Torino 10126, Italy
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy.,Institute of Crystallography, National Research Council of Italy, Via Amendola, 122/o, Bari 70126, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| |
Collapse
|
3
|
Jakowiecki J, Orzeł U, Gliździnska A, Możajew M, Filipek S. Specificities of Protein Homology Modeling for Allosteric Drug Design. Methods Mol Biol 2023; 2627:339-348. [PMID: 36959457 DOI: 10.1007/978-1-0716-2974-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
The allosteric binding sites are usually located in the flexible areas of proteins, which are hardly visible in the crystal structures. However, there are notable exceptions like allosteric sites in receptors in class B and C of GPCRs, which are located within a well-defined bundle of transmembrane helices. Class B and C evolved from class A and even after swapping of orthosteric and allosteric sites the central binding site persisted and it can be used for easy design of allosteric drugs. However, studying the ligand binding to the allosteric sites in the most populated class A of GPCRs is still a challenge, since they are located mostly in unresolved parts of the receptor's structure, and especially N-terminus. This chapter provides an example of cannabinoid CB1 receptor N-terminal homology modeling, ligand-guided modeling of the allosteric site in GABA receptor, as well as C-linker modeling in the potassium ion channels where the allosteric phospholipid ligand PIP2 is bound.
Collapse
Affiliation(s)
- Jakub Jakowiecki
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Urszula Orzeł
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Aleksandra Gliździnska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Mariusz Możajew
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Sławomir Filipek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
4
|
Mallo-Abreu A, Reyes-Resina I, Azuaje J, Franco R, García-Rey A, Majellaro M, Miranda-Pastoriza D, García-Mera X, Jespers W, Gutiérrez-de-Terán H, Navarro G, Sotelo E. Potent and Subtype-Selective Dopamine D 2 Receptor Biased Partial Agonists Discovered via an Ugi-Based Approach. J Med Chem 2021; 64:8710-8726. [PMID: 34110150 PMCID: PMC8552448 DOI: 10.1021/acs.jmedchem.1c00704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Using
a previously unexplored, efficient, and versatile multicomponent
method, we herein report the rapid generation of novel potent and
subtype-selective DRD2 biased partial agonists. This strategy
exemplifies the search for diverse and previously unexplored moieties
for the secondary/allosteric pharmacophore of the common phenyl-piperazine
scaffold. The pharmacological characterization of the new compound
series led to the identification of several ligands with excellent
DRD2 affinity and subtype selectivity and remarkable functional
selectivity for either the cAMP (22a and 24d) or the β-arrestin (27a and 29c)
signaling pathways. These results were further interpreted on the
basis of molecular models of these ligands in complex with the recent
DRD2 crystal structures, highlighting the critical role
of the secondary/allosteric pharmacophore in modulating the functional
selectivity profile.
Collapse
Affiliation(s)
- Ana Mallo-Abreu
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Jhonny Azuaje
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Rafael Franco
- Faculty of Chemistry, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Aitor García-Rey
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria Majellaro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Darío Miranda-Pastoriza
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Xerardo García-Mera
- Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Willem Jespers
- Department of Cell and Molecular Biology, Uppsala University, Uppsala SE-75124, Sweden
| | | | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Eddy Sotelo
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.,Departamento de Química Orgánica, Facultade de Farmacia, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Zhu S, Wu M, Huang Z, An J. Trends in application of advancing computational approaches in GPCR ligand discovery. Exp Biol Med (Maywood) 2021; 246:1011-1024. [PMID: 33641446 PMCID: PMC8113737 DOI: 10.1177/1535370221993422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) comprise the most important superfamily of protein targets in current ligand discovery and drug development. GPCRs are integral membrane proteins that play key roles in various cellular signaling processes. Therefore, GPCR signaling pathways are closely associated with numerous diseases, including cancer and several neurological, immunological, and hematological disorders. Computer-aided drug design (CADD) can expedite the process of GPCR drug discovery and potentially reduce the actual cost of research and development. Increasing knowledge of biological structures, as well as improvements on computer power and algorithms, have led to unprecedented use of CADD for the discovery of novel GPCR modulators. Similarly, machine learning approaches are now widely applied in various fields of drug target research. This review briefly summarizes the application of rising CADD methodologies, as well as novel machine learning techniques, in GPCR structural studies and bioligand discovery in the past few years. Recent novel computational strategies and feasible workflows are updated, and representative cases addressing challenging issues on olfactory receptors, biased agonism, and drug-induced cardiotoxic effects are highlighted to provide insights into future GPCR drug discovery.
Collapse
Affiliation(s)
- Siyu Zhu
- Division of Infectious Diseases and Global Public Health, Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Meixian Wu
- Division of Infectious Diseases and Global Public Health, Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Ziwei Huang
- Division of Infectious Diseases and Global Public Health, Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
- Ciechanover Institute of Precision and Regenerative Medicine, School of Life and Health Sciences, Chinese University of Hong Kong, Shenzhen 518172, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jing An
- Division of Infectious Diseases and Global Public Health, Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Mehta P, Miszta P, Filipek S. Molecular Modeling of Histamine Receptors-Recent Advances in Drug Discovery. Molecules 2021; 26:1778. [PMID: 33810008 PMCID: PMC8004658 DOI: 10.3390/molecules26061778] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
The recent developments of fast reliable docking, virtual screening and other algorithms gave rise to discovery of many novel ligands of histamine receptors that could be used for treatment of allergic inflammatory disorders, central nervous system pathologies, pain, cancer and obesity. Furthermore, the pharmacological profiles of ligands clearly indicate that these receptors may be considered as targets not only for selective but also for multi-target drugs that could be used for treatment of complex disorders such as Alzheimer's disease. Therefore, analysis of protein-ligand recognition in the binding site of histamine receptors and also other molecular targets has become a valuable tool in drug design toolkit. This review covers the period 2014-2020 in the field of theoretical investigations of histamine receptors mostly based on molecular modeling as well as the experimental characterization of novel ligands of these receptors.
Collapse
Affiliation(s)
| | | | - Sławomir Filipek
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland or (P.M.); (P.M.)
| |
Collapse
|
7
|
Désaubry L, Kanthasamy AG, Nebigil CG. Prokineticin signaling in heart-brain developmental axis: Therapeutic options for heart and brain injuries. Pharmacol Res 2020; 160:105190. [PMID: 32937177 PMCID: PMC7674124 DOI: 10.1016/j.phrs.2020.105190] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
Heart and brain development occur simultaneously during the embryogenesis, and both organ development and injuries are interconnected. Early neuronal and cardiac injuries share mutual cellular events, such as angiogenesis and plasticity that could either delay disease progression or, in the long run, result in detrimental health effects. For this reason, the common mechanisms provide a new and previously undervalued window of opportunity for intervention. Because angiogenesis, cardiogenesis and neurogenesis are essential for the development and regeneration of the heart and brain, we discuss therein the role of prokineticin as an angiogenic neuropeptide in heart-brain development and injuries. We focus on the role of prokineticin signaling and the effect of drugs targeting prokineticin receptors in neuroprotection and cardioprotection, with a special emphasis on heart failure, neurodegenerativParkinson's disease and ischemic heart and brain injuries. Indeed, prokineticin triggers common pro-survival signaling pathway in heart and brain. Our review aims at stimulating researchers and clinicians in neurocardiology to focus on the role of prokineticin signaling in the reciprocal interaction between heart and brain. We hope to facilitate the discovery of new treatment strategies, acting in both heart and brain degenerative diseases.
Collapse
Affiliation(s)
- Laurent Désaubry
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Canan G Nebigil
- Regenerative Nanomedicine, UMR 1260, INSERM, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
8
|
Tebo AG, Moeyaert B, Thauvin M, Carlon-Andres I, Böken D, Volovitch M, Padilla-Parra S, Dedecker P, Vriz S, Gautier A. Orthogonal fluorescent chemogenetic reporters for multicolor imaging. Nat Chem Biol 2020; 17:30-38. [PMID: 32778846 PMCID: PMC7610487 DOI: 10.1038/s41589-020-0611-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/02/2020] [Indexed: 12/24/2022]
Abstract
Spectrally separated fluorophores allow the observation of multiple targets simultaneously inside living cells, leading to a deeper understanding of the molecular interplay that regulates cell function and fate. Chemogenetic systems combining a tag and a synthetic fluorophore provide certain advantages over fluorescent proteins since there is no requirement for chromophore maturation. Here, we present the engineering of a set of spectrally orthogonal fluorogen activating tags based on the Fluorescence Activating and absorption Shifting Tag (FAST), that are compatible with two-color, live cell imaging. The resulting tags, greenFAST and redFAST, demonstrate orthogonality not only in their fluorogen recognition capabilities, but also in their one- and two-photon absorption profiles. This pair of orthogonal tags allowed the creation of a two-color cell cycle sensor capable of detecting very short, early cell cycles in zebrafish development, and the development of split complementation systems capable of detecting multiple protein-protein interactions by live cell fluorescence microscopy.
Collapse
Affiliation(s)
- Alison G Tebo
- Sorbonne University, École Normale Supérieure, PSL University, CNRS, Laboratoire des biomolécules (LBM), Paris, France.,PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne University, CNRS, Paris, France.,Janelia Farms Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Benjamien Moeyaert
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Marion Thauvin
- Center for Interdisciplinary Research, Collège de France, CNRS, INSERM, PSL University, Paris, France.,Sorbonne University, Paris, France
| | - Irene Carlon-Andres
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford, UK
| | - Dorothea Böken
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne University, CNRS, Paris, France
| | - Michel Volovitch
- Center for Interdisciplinary Research, Collège de France, CNRS, INSERM, PSL University, Paris, France.,Department of Biology, École Normale Supérieure, PSL University, Paris, France
| | - Sergi Padilla-Parra
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford, UK.,Department of Infectious Diseases, Faculty of Life Sciences & Medicine, King's College London, London, UK.,Randall Centre for Cell and Molecular Biology, King's College London, London, UK
| | - Peter Dedecker
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Sophie Vriz
- Center for Interdisciplinary Research, Collège de France, CNRS, INSERM, PSL University, Paris, France.,Faculty of Sciences, Université de Paris, Paris, France
| | - Arnaud Gautier
- Sorbonne University, École Normale Supérieure, PSL University, CNRS, Laboratoire des biomolécules (LBM), Paris, France. .,PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne University, CNRS, Paris, France. .,Institut Universitaire de France, Paris, France.
| |
Collapse
|
9
|
Chruścicka B, Wallace Fitzsimons SE, Borroto-Escuela DO, Druelle C, Stamou P, Nally K, Dinan TG, Cryan JF, Fuxe K, Schellekens H. Attenuation of Oxytocin and Serotonin 2A Receptor Signaling through Novel Heteroreceptor Formation. ACS Chem Neurosci 2019; 10:3225-3240. [PMID: 31038917 DOI: 10.1021/acschemneuro.8b00665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The oxytocin receptor (OTR) and the 5-hydroxytryptamine 2A receptor (5-HTR2A) are expressed in similar brain regions modulating central pathways critical for social and cognition-related behaviors. Signaling crosstalk between their endogenous ligands, oxytocin (OT) and serotonin (5-hydroxytryptamine, 5-HT), highlights the complex interplay between these two neurotransmitter systems and may be indicative of the formation of heteroreceptor complexes with subsequent downstream signaling changes. In this study, we assess the possible formation of OTR-5HTR2A heteromers in living cells and the functional downstream consequences of this receptor-receptor interaction. First, we demonstrated the existence of a physical interaction between the OTR and 5-HTR2A in vitro, using a flow cytometry-based FRET approach and confocal microscopy. Furthermore, we investigated the formation of this specific heteroreceptor complex ex vivo in the brain sections using the Proximity Ligation Assay (PLA). The OTR-5HTR2A heteroreceptor complexes were identified in limbic regions (including hippocampus, cingulate cortex, and nucleus accumbens), key regions associated with cognition and social-related behaviors. Next, functional cellular-based assays to assess the OTR-5HTR2A downstream signaling crosstalk showed a reduction in potency and efficacy of OT and OTR synthetic agonists, carbetocin and WAY267464, on OTR-mediated Gαq signaling. Similarly, the activation of 5-HTR2A by the endogenous agonist, 5-HT, also revealed attenuation in Gαq-mediated signaling. Finally, altered receptor trafficking within the cell was demonstrated, indicative of cotrafficking of the OTR/5-HTR2A pair. Overall, these results constitute a novel mechanism of specific interaction between the OT and 5-HT neurotransmitters via OTR-5HTR2A heteroreceptor formation and provide potential new therapeutic strategies in the treatment of social and cognition-related diseases.
Collapse
Affiliation(s)
- Barbara Chruścicka
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Shauna E. Wallace Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Clémentine Druelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Kenneth Nally
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockholm, Sweden
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
10
|
Analysis of a Protein Network Related to Copy Number Variations in Autism Spectrum Disorder. J Mol Neurosci 2019; 69:140-149. [DOI: 10.1007/s12031-019-01343-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 05/22/2019] [Indexed: 01/17/2023]
|
11
|
A light in the dark: state of the art and perspectives in optogenetics and optopharmacology for restoring vision. Future Med Chem 2019; 11:463-487. [DOI: 10.4155/fmc-2018-0315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the last decade, innovative therapeutic strategies against inherited retinal degenerations (IRDs) have emerged. In particular, chemical- and opto-genetics approaches or a combination of them have been identified for modulating neuronal/optical activity in order to restore vision in blinding diseases. The ‘chemical-genetics approach’ (optopharmacology) uses small molecules (exogenous photoswitches) for restoring light sensitivity by activating ion channels. The ‘opto-genetics approach’ employs light-activated photosensitive proteins (exogenous opsins), introduced by viral vectors in injured tissues, to restore light response. These approaches offer control of neuronal activities with spatial precision and limited invasiveness, although with some drawbacks. Currently, a combined therapeutic strategy (optogenetic pharmacology) is emerging. This review describes the state of the art and provides an overview of the future perspectives in vision restoration.
Collapse
|
12
|
Carli M, Kolachalam S, Aringhieri S, Rossi M, Giovannini L, Maggio R, Scarselli M. Dopamine D2 Receptors Dimers: How can we Pharmacologically Target Them? Curr Neuropharmacol 2018; 16:222-230. [PMID: 28521704 PMCID: PMC5883381 DOI: 10.2174/1570159x15666170518151127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/08/2017] [Accepted: 05/17/2017] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Dopamine D2 and D3 receptors can form homo- and heterodimers and are important targets in Schizophrenia and Parkinson's. Recently, many efforts have been made to pharmacologically target these receptor complexes. This review focuses on various strategies to act specifically on dopamine receptor dimers, that are transiently formed. METHODS Various binding and functional assays were reviewed to study the properties of bivalent ligands, particularly for the dualsteric compound SB269,652. The dimerization of D2 and D3 receptors were analyzed by using single particle tracking microscopy. RESULTS The specific targeting of dopamine D2 and D3 dimers can be achieved with bifunctional ligands, composed of two pharmacophores binding the two orthosteric sites of the dimeric complex. If the target is a homodimer, then the ligand is homobivalent. Instead, if the target is a heterodimer, then the ligand is heterobivalent. However, there is some concern regarding pharmacokinetics and binding properties of such drugs. Recently, a new generation of bitopic compounds with dualsteric properties have been discovered that bind to the orthosteric and the allosteric sites in one monomeric receptor. Regarding dopamine D2 and D3 receptors, a new dualsteric molecule SB269,652 was shown to have selective negative allosteric properties across D2 and D3 homodimers, but it behaves as an orthosteric antagonist on receptor monomer. Targeting dimers is also complicated as they are transiently formed with varying monomer/dimer ratio. Furthermore, this ratio can be altered by administering an agonist or a bifunctional antagonist. CONCLUSION Last 15 years have witnessed an explosive amount of work aimed at generating bifunctional compounds as a novel strategy to target GPCR homo- and heterodimers, including dopamine receptors. Their clinical use is far from trivial, but, at least, they have been used to validate the existence of receptor dimers in-vitro and in-vivo. The dualsteric compound SB269, 652, with its peculiar pharmacological profile, may offer therapeutic advantages and a better tolerability in comparison with pure antagonists at D2 and D3 receptors and pave the way for a new generation of antipsychotic drugs.
Collapse
Affiliation(s)
- Marco Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Stefano Aringhieri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD. United States
| | - Luca Giovannini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| |
Collapse
|
13
|
Singh SS, Jois SD. Homo- and Heterodimerization of Proteins in Cell Signaling: Inhibition and Drug Design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 111:1-59. [PMID: 29459028 DOI: 10.1016/bs.apcsb.2017.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Protein dimerization controls many physiological processes in the body. Proteins form homo-, hetero-, or oligomerization in the cellular environment to regulate the cellular processes. Any deregulation of these processes may result in a disease state. Protein-protein interactions (PPIs) can be inhibited by antibodies, small molecules, or peptides, and inhibition of PPI has therapeutic value. PPI drug discovery research has steadily increased in the last decade, and a few PPI inhibitors have already reached the pharmaceutical market. Several PPI inhibitors are in clinical trials. With advancements in structural and molecular biology methods, several methods are now available to study protein homo- and heterodimerization and their inhibition by drug-like molecules. Recently developed methods to study PPI such as proximity ligation assay and enzyme-fragment complementation assay that detect the PPI in the cellular environment are described with examples. At present, the methods used to design PPI inhibitors can be classified into three major groups: (1) structure-based drug design, (2) high-throughput screening, and (3) fragment-based drug design. In this chapter, we have described some of the experimental methods to study PPIs and their inhibition. Examples of homo- and heterodimers of proteins, their structural and functional aspects, and some of the inhibitors that have clinical importance are discussed. The design of PPI inhibitors of epidermal growth factor receptor heterodimers and CD2-CD58 is discussed in detail.
Collapse
Affiliation(s)
- Sitanshu S Singh
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Seetharama D Jois
- Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States.
| |
Collapse
|
14
|
Lee Y, Basith S, Choi S. Recent Advances in Structure-Based Drug Design Targeting Class A G Protein-Coupled Receptors Utilizing Crystal Structures and Computational Simulations. J Med Chem 2017; 61:1-46. [PMID: 28657745 DOI: 10.1021/acs.jmedchem.6b01453] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) represent the largest and most physiologically important integral membrane protein family, and these receptors respond to a wide variety of physiological and environmental stimuli. GPCRs are among the most critical therapeutic targets for numerous human diseases, and approximately one-third of the currently marketed drugs target this receptor family. The recent breakthroughs in GPCR structural biology have significantly contributed to our understanding of GPCR function, ligand binding, and pharmacological action as well as to the design of new drugs. This perspective highlights the latest advances in GPCR structures with a focus on the receptor-ligand interactions of each receptor family in class A nonrhodopsin GPCRs as well as the structural features for their activation, biased signaling, and allosteric mechanisms. The current state-of-the-art methodologies of structure-based drug design (SBDD) approaches in the GPCR research field are also discussed.
Collapse
Affiliation(s)
- Yoonji Lee
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| | - Shaherin Basith
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| | - Sun Choi
- National Leading Research Laboratory (NLRL) of Molecular Modeling & Drug Design, College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul 03760, Republic of Korea
| |
Collapse
|
15
|
Lifshitz V, Priceman SJ, Li W, Cherryholmes G, Lee H, Makovski-Silverstein A, Borriello L, DeClerck YA, Yu H. Sphingosine-1-Phosphate Receptor-1 Promotes Environment-Mediated and Acquired Chemoresistance. Mol Cancer Ther 2017; 16:2516-2527. [PMID: 28716816 DOI: 10.1158/1535-7163.mct-17-0379] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/09/2017] [Accepted: 07/05/2017] [Indexed: 01/05/2023]
Abstract
Drug resistance is a major barrier for the development of effective and durable cancer therapies. Overcoming this challenge requires further defining the cellular and molecular mechanisms underlying drug resistance, both acquired and environment-mediated drug resistance (EMDR). Here, using neuroblastoma (NB), a childhood cancer with high incidence of recurrence due to resistance to chemotherapy, as a model we show that human bone marrow-mesenchymal stromal cells induce tumor expression of sphingosine-1-phosphate receptor-1 (S1PR1), leading to their resistance to chemotherapy. Targeting S1PR1 by shRNA markedly enhances etoposide-induced apoptosis in NB cells and abrogates EMDR, while overexpression of S1PR1 significantly protects NB cells from multidrug-induced apoptosis via activating JAK-STAT3 signaling. Elevated S1PR1 expression and STAT3 activation are also observed in human NB cells with acquired resistance to etoposide. We show in vitro and in human NB xenograft models that treatment with FTY720, an FDA-approved drug and antagonist of S1PR1, dramatically sensitizes drug-resistant cells to etoposide. In summary, we identify S1PR1 as a critical target for reducing both EMDR and acquired chemoresistance in NB. Mol Cancer Ther; 16(11); 2516-27. ©2017 AACR.
Collapse
Affiliation(s)
- Veronica Lifshitz
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Saul J Priceman
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California.
| | - Wenzhao Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Gregory Cherryholmes
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Heehyoung Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Adar Makovski-Silverstein
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Lucia Borriello
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California
| | - Yves A DeClerck
- Division of Hematology, Oncology and Blood and Bone Marrow Transplantation, Department of Pediatrics, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, California. .,Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California
| | - Hua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
16
|
Cordier D, Krolicki L, Morgenstern A, Merlo A. Targeted Radiolabeled Compounds in Glioma Therapy. Semin Nucl Med 2016; 46:243-9. [PMID: 27067505 DOI: 10.1053/j.semnuclmed.2016.01.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Malignant gliomas of World Health Organization (WHO) grades II-IV represent the largest entity within the group of intrinsic brain tumors and are graded according to their pathophysiological features with survival times between more than 10 years (WHO II) and only several months (WHO IV). Gliomas arise from astrocytic or oligodendrocytic precursor cells and exhibit an infiltrative growth pattern lacking a clearly identifiable tumor border. The development of effective treatment strategies of the invasive tumor cell front represents the main challenge in glioma therapy. The therapeutic standard consists of surgical resection and, depending on the extent of resection and WHO grade, adjuvant external beam radiotherapy or systemic chemotherapy. Within the last decades, there has been no major improvement of the prognosis of patients with glioma. The consistent overexpression of neurokinin type 1 receptors in gliomas WHO grades II-IV has been used to develop a therapeutic substance P-based targeting system. A substance P-analogue conjugated to the DOTA or DOTAGA chelator has been labeled with different alpha-particle or beta-particle emitting radionuclides for targeted glioma therapy. The radiopharmaceutical has been locally injected into the tumors or the resection cavity. In several clinical studies, the methodology has been examined in adjuvant and neoadjuvant clinical settings. Although no large controlled series have so far been generated, the results of radiolabeled substance P-based targeted glioma therapy compare favorably with standard therapy. Recently, labeling with the alpha particle emitting Bi-213 has been found to be promising due to the high linear energy transfer and the very short tissue range of 0.08 mm. Further development needs to focus on the improvement of the stability of the compound and the application by dedicated catheter systems to improve the intratumoral distribution of the radiopharmaceutical within the prognostically critical infiltrative growing zone of the glioma.
Collapse
Affiliation(s)
- Dominik Cordier
- Department of Neurosurgery, University of Basel, Basel, Switzerland
| | - Leszek Krolicki
- Department of Nuclear Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Institute for Transuranium Elements, Karlsruhe, Germany
| | - Adrian Merlo
- Department of Neurosurgery, University of Basel, Basel, Switzerland.
| |
Collapse
|
17
|
Maramai S, Gemma S, Brogi S, Campiani G, Butini S, Stark H, Brindisi M. Dopamine D3 Receptor Antagonists as Potential Therapeutics for the Treatment of Neurological Diseases. Front Neurosci 2016; 10:451. [PMID: 27761108 PMCID: PMC5050208 DOI: 10.3389/fnins.2016.00451] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/20/2016] [Indexed: 01/09/2023] Open
Abstract
D3 receptors represent a major focus of current drug design and development of therapeutics for dopamine-related pathological states. Their close homology with the D2 receptor subtype makes the development of D3 selective antagonists a challenging task. In this review, we explore the relevance and therapeutic utility of D3 antagonists or partial agonists endowed with multireceptor affinity profile in the field of central nervous system disorders such as schizophrenia and drug abuse. In fact, the peculiar distribution and low brain abundance of D3 receptors make them a valuable target for the development of drugs devoid of motor side effects classically elicited by D2 antagonists. Recent research efforts were devoted to the conception of chemical templates possibly endowed with a multi-target profile, especially with regards to other G-protein-coupled receptors (GPCRs). A comprehensive overview of the recent literature in the field is herein provided. In particular, the evolution of the chemical templates has been tracked, according to the growing advancements in both the structural information and the refinement of the key pharmacophoric elements. The receptor/multireceptor affinity and functional profiles for the examined compounds have been covered, together with their most significant pharmacological applications.
Collapse
Affiliation(s)
- Samuele Maramai
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Simone Brogi
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Stefania Butini
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| | - Holger Stark
- Institut fuer Pharmazeutische and Medizinische Chemie, Heinrich-Heine-Universitaet Duesseldorf Duesseldorf, Germany
| | - Margherita Brindisi
- European Research Centre for Drug Discovery and Development and Department of Biotechnology, Chemistry and Pharmacy, University of Siena Siena, Italy
| |
Collapse
|
18
|
Arreola R, Alvarez-Herrera S, Pérez-Sánchez G, Becerril-Villanueva E, Cruz-Fuentes C, Flores-Gutierrez EO, Garcés-Alvarez ME, de la Cruz-Aguilera DL, Medina-Rivero E, Hurtado-Alvarado G, Quintero-Fabián S, Pavón L. Immunomodulatory Effects Mediated by Dopamine. J Immunol Res 2016; 2016:3160486. [PMID: 27795960 PMCID: PMC5067323 DOI: 10.1155/2016/3160486] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/29/2016] [Accepted: 08/08/2016] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA), a neurotransmitter in the central nervous system (CNS), has modulatory functions at the systemic level. The peripheral and central nervous systems have independent dopaminergic system (DAS) that share mechanisms and molecular machinery. In the past century, experimental evidence has accumulated on the proteins knowledge that is involved in the synthesis, reuptake, and transportation of DA in leukocytes and the differential expression of the D1-like (D1R and D5R) and D2-like receptors (D2R, D3R, and D4R). The expression of these components depends on the state of cellular activation and the concentration and time of exposure to DA. Receptors that are expressed in leukocytes are linked to signaling pathways that are mediated by changes in cAMP concentration, which in turn triggers changes in phenotype and cellular function. According to the leukocyte lineage, the effects of DA are associated with such processes as respiratory burst, cytokine and antibody secretion, chemotaxis, apoptosis, and cytotoxicity. In clinical conditions such as schizophrenia, Parkinson disease, Tourette syndrome, and multiple sclerosis (MS), there are evident alterations during immune responses in leukocytes, in which changes in DA receptor density have been observed. Several groups have proposed that these findings are useful in establishing clinical status and clinical markers.
Collapse
Affiliation(s)
- Rodrigo Arreola
- Psychiatric Genetics Department, National Institute of Psychiatry “Ramón de la Fuente”, Clinical Research Branch, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Samantha Alvarez-Herrera
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Gilberto Pérez-Sánchez
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Enrique Becerril-Villanueva
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Carlos Cruz-Fuentes
- Psychiatric Genetics Department, National Institute of Psychiatry “Ramón de la Fuente”, Clinical Research Branch, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Enrique Octavio Flores-Gutierrez
- National Institute of Psychiatry “Ramón de la Fuente”, Clinical Research Branch, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - María Eugenia Garcés-Alvarez
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| | - Dora Luz de la Cruz-Aguilera
- Laboratory of Neuroimmunoendocrinology, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Avenida Insurgentes Sur 3877, La Fama, Tlalpan, 14269 Mexico City, Mexico
| | - Emilio Medina-Rivero
- Unidad de Investigación y Desarrollo, Probiomed S.A. de C.V. Cruce de Carreteras Acatzingo-Zumpahuacán S/N, 52400 Tenancingo, MEX, Mexico
| | - Gabriela Hurtado-Alvarado
- Area of Neurosciences, Department of Biology of Reproduction, CBS, Universidad Autonoma Metropolitana, Unidad Iztapalapa, Avenida San Rafael Atlixco No. 186, Colonia Vicentina, Iztapalapa, 09340 Mexico City, Mexico
| | - Saray Quintero-Fabián
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Instituto Nacional de Pediatría, Av. del Iman No. 1, Cuarto Piso, 04530 Mexico City, Mexico
| | - Lenin Pavón
- Department of Psychoimmunology, National Institute of Psychiatry “Ramón de la Fuente”, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, 14370 Mexico City, Mexico
| |
Collapse
|
19
|
Nemet I, Ropelewski P, Imanishi Y. Applications of phototransformable fluorescent proteins for tracking the dynamics of cellular components. Photochem Photobiol Sci 2016; 14:1787-806. [PMID: 26345171 DOI: 10.1039/c5pp00174a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the past few decades, fluorescent proteins have revolutionized the field of cell biology. Phototransformable fluorescent proteins are capable of changing their excitation and emission spectra after being exposed to specific wavelength(s) of light. The majority of phototransformable fluorescent proteins have originated from marine organisms. Genetic engineering of these proteins has made available many choices for different colors, modes of conversion, and other biophysical properties. Their phototransformative property has allowed the highlighting and tracking of subpopulations of cells, organelles, and proteins in living systems. Furthermore, phototransformable fluorescent proteins have offered new methods for superresolution fluorescence microscopy and optogenetics manipulation of proteins. One of the major advantages of phototransformable fluorescent proteins is their applicability for visualizing newly synthesized proteins that are en route to their final destinations. In this paper, we will discuss the biological applications of phototransformable fluorescent proteins with special emphasis on the application of tracking membrane proteins in vertebrate photoreceptor cells.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
20
|
Homogeneous time-resolved G protein-coupled receptor–ligand binding assay based on fluorescence cross-correlation spectroscopy. Anal Biochem 2016; 502:24-35. [DOI: 10.1016/j.ab.2016.02.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/19/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
|
21
|
Butini S, Nikolic K, Kassel S, Brückmann H, Filipic S, Agbaba D, Gemma S, Brogi S, Brindisi M, Campiani G, Stark H. Polypharmacology of dopamine receptor ligands. Prog Neurobiol 2016; 142:68-103. [PMID: 27234980 DOI: 10.1016/j.pneurobio.2016.03.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 01/26/2016] [Accepted: 03/15/2016] [Indexed: 01/11/2023]
Abstract
Most neurological diseases have a multifactorial nature and the number of molecular mechanisms discovered as underpinning these diseases is continuously evolving. The old concept of developing selective agents for a single target does not fit with the medical need of most neurological diseases. The development of designed multiple ligands holds great promises and appears as the next step in drug development for the treatment of these multifactorial diseases. Dopamine and its five receptor subtypes are intimately involved in numerous neurological disorders. Dopamine receptor ligands display a high degree of cross interactions with many other targets including G-protein coupled receptors, transporters, enzymes and ion channels. For brain disorders like Parkinsońs disease, schizophrenia and depression the dopaminergic system, being intertwined with many other signaling systems, plays a key role in pathogenesis and therapy. The concept of designed multiple ligands and polypharmacology, which perfectly meets the therapeutic needs for these brain disorders, is herein discussed as a general ligand-based concept while focusing on dopaminergic agents and receptor subtypes in particular.
Collapse
Affiliation(s)
- S Butini
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - K Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - S Kassel
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - H Brückmann
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - S Filipic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - D Agbaba
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - S Gemma
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - S Brogi
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - M Brindisi
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - G Campiani
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - H Stark
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany.
| |
Collapse
|
22
|
Allegretti M, Cesta MC, Locati M. Allosteric Modulation of Chemoattractant Receptors. Front Immunol 2016; 7:170. [PMID: 27199992 PMCID: PMC4852175 DOI: 10.3389/fimmu.2016.00170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/18/2016] [Indexed: 01/05/2023] Open
Abstract
Chemoattractants control selective leukocyte homing via interactions with a dedicated family of related G protein-coupled receptor (GPCR). Emerging evidence indicates that the signaling activity of these receptors, as for other GPCR, is influenced by allosteric modulators, which interact with the receptor in a binding site distinct from the binding site of the agonist and modulate the receptor signaling activity in response to the orthosteric ligand. Allosteric modulators have a number of potential advantages over orthosteric agonists/antagonists as therapeutic agents and offer unprecedented opportunities to identify extremely selective drug leads. Here, we resume evidence of allosterism in the context of chemoattractant receptors, discussing in particular its functional impact on functional selectivity and probe/concentration dependence of orthosteric ligands activities.
Collapse
Affiliation(s)
| | | | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Segrate, Italy; Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
23
|
Franco R, Martínez-Pinilla E, Lanciego JL, Navarro G. Basic Pharmacological and Structural Evidence for Class A G-Protein-Coupled Receptor Heteromerization. Front Pharmacol 2016; 7:76. [PMID: 27065866 PMCID: PMC4815248 DOI: 10.3389/fphar.2016.00076] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/11/2016] [Indexed: 12/25/2022] Open
Abstract
Cell membrane receptors rarely work on isolation, often they form oligomeric complexes with other receptor molecules and they may directly interact with different proteins of the signal transduction machinery. For a variety of reasons, rhodopsin-like class A G-protein-coupled receptors (GPCRs) seem an exception to the general rule of receptor-receptor direct interaction. In fact, controversy surrounds their potential to form homo- hetero-dimers/oligomers with other class A GPCRs; in a sense, the field is going backward instead of forward. This review focuses on the convergent, complementary and telling evidence showing that homo- and heteromers of class A GPCRs exist in transfected cells and, more importantly, in natural sources. It is time to decide between questioning the occurrence of heteromers or, alternatively, facing the vast scientific and technical challenges that class A receptor-dimer/oligomer existence pose to Pharmacology and to Drug Discovery.
Collapse
Affiliation(s)
- Rafael Franco
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Universitat de BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos IIIMadrid, Spain; Institute of Biomedicine, University of BarcelonaBarcelona, Spain
| | - Eva Martínez-Pinilla
- Instituto de Neurociencias del Principado de Asturias, Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de OviedoAsturias, Spain; Neurosciences Division, Centre for Applied Medical Research, University of NavarraPamplona, Spain; Instituto de Investigaciones Sanitarias de NavarraPamplona, Spain
| | - José L Lanciego
- Centro de Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos IIIMadrid, Spain; Neurosciences Division, Centre for Applied Medical Research, University of NavarraPamplona, Spain; Instituto de Investigaciones Sanitarias de NavarraPamplona, Spain
| | - Gemma Navarro
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Universitat de BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos IIIMadrid, Spain
| |
Collapse
|
24
|
Abstract
G protein-coupled receptors (GPCRs) remain a major domain of pharmaceutical discovery. The identification of GPCR lead compounds and their optimization are now structure-based, thanks to advances in X-ray crystallography, molecular modeling, protein engineering and biophysical techniques. In silico screening provides useful hit molecules. New pharmacological approaches to tuning the pleotropic action of GPCRs include: allosteric modulators, biased ligands, GPCR heterodimer-targeted compounds, manipulation of polypharmacology, receptor antibodies and tailoring of drug molecules to fit GPCR pharmacogenomics. Measurements of kinetics and drug efficacy are factors influencing clinical success. With the exception of inhibitors of GPCR kinases, targeting of intracellular GPCR signaling or receptor cycling for therapeutic purposes remains a futuristic concept. New assay approaches are more efficient and multidimensional: cell-based, label-free, fluorescence-based assays, and biosensors. Tailoring GPCR drugs to a patient's genetic background is now being considered. Chemoinformatic tools can predict ADME-tox properties. New imaging technology visualizes drug action in vivo. Thus, there is reason to be optimistic that new technology for GPCR ligand discovery will help reverse the current narrowing of the pharmaceutical pipeline.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, Maryland 20892, USA.
| |
Collapse
|
25
|
Massotte D. Editorial: Monitoring endogenous GPCRs: lessons for drug design. Front Pharmacol 2015; 6:146. [PMID: 26236238 PMCID: PMC4500908 DOI: 10.3389/fphar.2015.00146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/02/2022] Open
Affiliation(s)
- Dominique Massotte
- Institut des Neurosciences Cellulaires et Intégratives Strasbourg, France
| |
Collapse
|
26
|
Park F. Activators of G protein signaling in the kidney. J Pharmacol Exp Ther 2015; 353:235-45. [PMID: 25628392 PMCID: PMC4407716 DOI: 10.1124/jpet.115.222695] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/23/2015] [Indexed: 12/15/2022] Open
Abstract
Heterotrimeric G proteins play a crucial role in regulating signal processing to maintain normal cellular homeostasis, and subtle perturbations in its activity can potentially lead to the pathogenesis of renal disorders or diseases. Cell-surface receptors and accessory proteins, which normally modify and organize the coupling of individual G protein subunits, contribute to the regulation of heterotrimeric G protein activity and their convergence and/or divergence of downstream signaling initiated by effector systems. Activators of G protein signaling (AGS) are a family of accessory proteins that intervene at multiple distinct points during the activation-inactivation cycle of G proteins, even in the absence of receptor stimulation. Perturbations in the expression of individual AGS proteins have been reported to modulate signal transduction pathways in a wide array of diseases and disorders within the brain, heart, immune system, and more recently, the kidney. This review will provide an overview of the expression profile, localization, and putative biologic role of the AGS family in the context of normal and diseased states of the kidney.
Collapse
Affiliation(s)
- Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|