1
|
Sharrocks KL, Swaih AM, Hanyaloglu AC. Single-molecule localization microscopy as a tool to quantify di/oligomerization of receptor tyrosine kinases and G protein-coupled receptors. Mol Pharmacol 2025; 107:100033. [PMID: 40228395 DOI: 10.1016/j.molpha.2025.100033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Dimerization and oligomerization of membrane receptors, including G protein-coupled receptors and receptor tyrosine kinases, are fundamental for regulating cell signaling and diversifying downstream responses to mediate a range of physiological processes. Receptor di/oligomers play roles in diverse facets of receptor function. Changes in receptor di/oligomers have been implicated in a range of diseases; therefore, better understanding of the specific composition and interactions between receptors in complexes is essential, especially for the development of di/oligomer-specific drugs. Previously, different optical microscopy approaches and proximity-based biophysical assays have been used to demonstrate di/oligomerization of membrane receptors. However, in recent years, single-molecule super-resolution microscopy techniques have allowed researchers to quantify and uncover the precise dynamics and stoichiometry of specific receptor complexes. This allows the organization of membrane protein receptors to be mapped across the plasma membrane to explore the effects of factors such as ligands, effectors, membrane environment, and therapeutic agents. Quantification of receptor complexes is required to better understand the intricate balance of distinct receptor complexes in cells. In this brief review, we provide an overview of single-molecule approaches for the quantification of receptor di/oligomerization. We will discuss the techniques commonly employed to study membrane receptor di/oligomerization and their relative advantages and limitations. SIGNIFICANCE STATEMENT: Receptor di/oligomerization plays an important role in their function. For some receptors, di/oligomerization is essential for functional signaling, whereas for others, it acts as a mechanism to achieve signaling pleiotropy. Aberrant receptor di/oligomerization has been implicated in a wide range of diseases. Single-molecule super-resolution microscopy techniques provide convincing methods to precisely quantify receptor complexes at the plasma membrane. Understanding receptor complex organization in disease models can also influence the targeting of specific monomeric or oligomeric complexes in therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Aylin C Hanyaloglu
- The Francis Crick Institute, London, UK; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
2
|
Wang Y, Wei L, Tan M, Yang Z, Gao B, Li J, Liu Y, Zikereya T, Shi K, Chen W. Aerobic exercise improves motor dysfunction in Parkinson's model mice via differential regulation of striatal medium spiny neuron. Sci Rep 2024; 14:12132. [PMID: 38802497 PMCID: PMC11130133 DOI: 10.1038/s41598-024-63045-4] [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: 02/04/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
The striatum plays a crucial role in providing input to the basal ganglia circuit and is implicated in the pathological process of Parkinson's disease (PD). Disruption of the dynamic equilibrium in the basal ganglia loop can be attributed to the abnormal functioning of the medium spiny neurons (MSNs) within the striatum, potentially acting as a trigger for PD. Exercise has been shown to mitigate striatal neuronal dysfunction through neuroprotective and neurorestorative effects and to improve behavioral deficits in PD model mice. In addition, this effect is offset by the activation of MSNs expressing dopamine D2 receptors (D2-MSNs). In the current study, we investigated the underlying neurobiological mechanisms of this effect. Our findings indicated that exercise reduces the power spectral density of the beta-band in the striatum and decreases the overall firing frequency of MSNs, particularly in the case of striatal D2-MSNs. These observations were consistent with the results of molecular biology experiments, which revealed that aerobic training specifically enhanced the expression of striatal dopamine D2 receptors (D2R). Taken together, our results suggest that aerobic training aimed at upregulating striatal D2R expression to inhibit the functional activity of D2-MSNs represents a potential therapeutic strategy for the amelioration of motor dysfunction in PD.
Collapse
Affiliation(s)
- Yinhao Wang
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Longwei Wei
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Mingli Tan
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Zizheng Yang
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Bo Gao
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Juan Li
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Yang Liu
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China
| | - Talifu Zikereya
- Department of Physical Education, China University of Geoscience, Beijing, China
| | - Kaixuan Shi
- Department of Physical Education, China University of Geoscience, Beijing, China.
| | - Wei Chen
- School of Physical Education, Hebei Normal University, Shijiazhuang, China.
- Key Laboratory of Measurement and Evaluation in Exercise Bioinformation of Hebei Province, Shijiazhuang, China.
| |
Collapse
|
3
|
de Bem Alves AC, Aguiar AS. Caffeine plus haloperidol reduces fatigue in an experimental model of Parkinson's disease - a prospective to A 2AR-D 2R heterodimer antagonism. Purinergic Signal 2024; 20:29-34. [PMID: 36918462 PMCID: PMC10828253 DOI: 10.1007/s11302-023-09933-2] [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: 11/17/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Fatigue is a non-motor symptom of Parkinson's disease (PD). Adenosine 2A receptor (A2AR) and compromised dopamine neurotransmission are linked to fatigue. Studies demonstrate that A2AR antagonism potentiates dopamine transmission via dopamine receptor D2 (D2R). However, the heterodimer form of A2AR-D2R in the striatum prompted questions about the therapeutic targets for PD patients. This study investigates the effects of caffeine (A2AR non-selective antagonist) plus haloperidol (D2R selective antagonist) treatment in the fatigue induced by the reserpine model of PD. Reserpinized mice showed impaired motor control in the open field test (p < 0.05) and fatigue in the grip strength meter test (p < 0.05). L-DOPA and caffeine plus haloperidol similarly increased motor control (p < 0.05) and mitigated fatigue (p < 0.05). Our results support the A2AR-D2R heterodimer participation in the central fatigue of PD, and highlight the potential of A2AR-D2R antagonism in the management of PD.
Collapse
Affiliation(s)
- Ana Cristina de Bem Alves
- Laboratory of Exercise Biology (LaBioEx), Department of Health Sciences, Federal University of Santa Catarina (UFSC), Ararangua, SC, 88905-120, Brazil.
| | - Aderbal Silva Aguiar
- Laboratory of Exercise Biology (LaBioEx), Department of Health Sciences, Federal University of Santa Catarina (UFSC), Ararangua, SC, 88905-120, Brazil
| |
Collapse
|
4
|
Jenner P, Falup-Pecurariu C, Leta V, Verin M, Auffret M, Bhidayasiri R, Weiss D, Borovečki F, Jost WH. Adopting the Rumsfeld approach to understanding the action of levodopa and apomorphine in Parkinson's disease. J Neural Transm (Vienna) 2023; 130:1337-1347. [PMID: 37210460 PMCID: PMC10645644 DOI: 10.1007/s00702-023-02655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/14/2023] [Indexed: 05/22/2023]
Abstract
Dopaminergic therapies dominate the treatment of the motor and non-motor symptoms of Parkinson's disease (PD) but there have been no major advances in therapy in many decades. Two of the oldest drugs used appear more effective than others-levodopa and apomorphine-but the reasons for this are seldom discussed and this may be one cause for a lack of progress. This short review questions current thinking on drug action and looks at whether adopting the philosophy of ex-US Secretary of State Donald Rumsfeld reveals 'unknown' aspects of the actions of levodopa and apomorphine that provide clues for a way forward. It appears that both levodopa and apomorphine have a more complex pharmacology than classical views would suggest. In addition, there are unexpected facets to the mechanisms through which levodopa acts that are either forgotten as 'known unknowns' or ignored as 'unknown unknowns'. The conclusion reached is that we may not know as much as we think about drug action in PD and there is a case for looking beyond the obvious.
Collapse
Affiliation(s)
- P Jenner
- Faculty of Life Sciences and Medicine, Institute of Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
| | - C Falup-Pecurariu
- Department of Neurology, Transylvania University, 500036, Brasov, Romania
| | - V Leta
- Parkinson's Foundation Center of Excellence at King's College Hospital; Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London and National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre, Institute of Psychology, Psychiatry and Neurosciences, King's College London, London, UK
| | - M Verin
- Institut des Neurosciences Cliniques de Rennes (INCR); Behavior and Basal Ganglia Research Unit, CIC-IT, CIC1414, Pontchaillou University Hospital and University of Rennes, Rennes, France
| | - M Auffret
- Institut des Neurosciences Cliniques de Rennes (INCR); Behavior and Basal Ganglia Research Unit, CIC-IT, CIC1414, Pontchaillou University Hospital and University of Rennes, Rennes, France
- France Développement Electronique (FDE), Monswiller, France
| | - Roongroj Bhidayasiri
- Department of Medicine, Faculty of Medicine, Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, 10330, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, 10330, Thailand
| | - D Weiss
- Department for Neurodegenerative Diseases, Centre for Neurology, Hertie-Institute for Clinical Brain Research, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - F Borovečki
- Division for Neurodegenerative Diseases and Neurogenomics, Department of Neurology, University Hospital Centre Zagreb, 10000, Zagreb, Croatia
| | - W H Jost
- Parkinson-Klinik Ortenau, Kreuzbergstr. 12-16, 77709, Wolfach, Germany
| |
Collapse
|
5
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, De Caro R, Maura G, Agnati LF. Modulation of Neuron and Astrocyte Dopamine Receptors via Receptor-Receptor Interactions. Pharmaceuticals (Basel) 2023; 16:1427. [PMID: 37895898 PMCID: PMC10610355 DOI: 10.3390/ph16101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Dopamine neurotransmission plays critical roles in regulating complex cognitive and behavioral processes including reward, motivation, reinforcement learning, and movement. Dopamine receptors are classified into five subtypes, widely distributed across the brain, including regions responsible for motor functions and specific areas related to cognitive and emotional functions. Dopamine also acts on astrocytes, which express dopamine receptors as well. The discovery of direct receptor-receptor interactions, leading to the formation of multimeric receptor complexes at the cell membrane and providing the cell decoding apparatus with flexible dynamics in terms of recognition and signal transduction, has expanded the knowledge of the G-protein-coupled receptor-mediated signaling processes. The purpose of this review article is to provide an overview of currently identified receptor complexes containing dopamine receptors and of their modulatory action on dopamine-mediated signaling between neurons and between neurons and astrocytes. Pharmacological possibilities offered by targeting receptor complexes in terms of addressing neuropsychiatric disorders associated with altered dopamine signaling will also be briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Cinzia Tortorella
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Manuela Marcoli
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Chiara Cervetto
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Raffaele De Caro
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Guido Maura
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| |
Collapse
|
6
|
Vellucci L, Ciccarelli M, Buonaguro EF, Fornaro M, D’Urso G, De Simone G, Iasevoli F, Barone A, de Bartolomeis A. The Neurobiological Underpinnings of Obsessive-Compulsive Symptoms in Psychosis, Translational Issues for Treatment-Resistant Schizophrenia. Biomolecules 2023; 13:1220. [PMID: 37627285 PMCID: PMC10452784 DOI: 10.3390/biom13081220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Almost 25% of schizophrenia patients suffer from obsessive-compulsive symptoms (OCS) considered a transdiagnostic clinical continuum. The presence of symptoms pertaining to both schizophrenia and obsessive-compulsive disorder (OCD) may complicate pharmacological treatment and could contribute to lack or poor response to the therapy. Despite the clinical relevance, no reviews have been recently published on the possible neurobiological underpinnings of this comorbidity, which is still unclear. An integrative view exploring this topic should take into account the following aspects: (i) the implication for glutamate, dopamine, and serotonin neurotransmission as demonstrated by genetic findings; (ii) the growing neuroimaging evidence of the common brain regions and dysfunctional circuits involved in both diseases; (iii) the pharmacological modulation of dopaminergic, serotoninergic, and glutamatergic systems as current therapeutic strategies in schizophrenia OCS; (iv) the recent discovery of midbrain dopamine neurons and dopamine D1- and D2-like receptors as orchestrating hubs in repetitive and psychotic behaviors; (v) the contribution of N-methyl-D-aspartate receptor subunits to both psychosis and OCD neurobiology. Finally, we discuss the potential role of the postsynaptic density as a structural and functional hub for multiple molecular signaling both in schizophrenia and OCD pathophysiology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry University Medical School of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
7
|
Cervetto C, Maura G, Guidolin D, Amato S, Ceccoli C, Agnati LF, Marcoli M. Striatal astrocytic A2A-D2 receptor-receptor interactions and their role in neuropsychiatric disorders. Neuropharmacology 2023:109636. [PMID: 37321323 DOI: 10.1016/j.neuropharm.2023.109636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 06/17/2023]
Abstract
It is now generally accepted that astrocytes are active players in synaptic transmission, so that a neurocentric perspective of the integrative signal communication in the central nervous system is shifting towards a neuro-astrocentric perspective. Astrocytes respond to synaptic activity, release chemical signals (gliotransmitters) and express neurotransmitter receptors (G protein-coupled and ionotropic receptors), thus behaving as co-actors with neurons in signal communication in the central nervous system. The ability of G protein-coupled receptors to physically interact through heteromerization, forming heteromers and receptor mosaics with new distinct signal recognition and transduction pathways, has been intensively studied at neuronal plasma membrane, and has changed the view of the integrative signal communication in the central nervous system. One of the best-known examples of receptor-receptor interaction through heteromerization, with relevant consequences for both the physiological and the pharmacological points of view, is given by adenosine A2A and dopamine D2 receptors on the plasma membrane of striatal neurons. Here we review evidence that native A2A and D2 receptors can interact through heteromerization at the plasma membrane of astrocytes as well. Astrocytic A2A-D2 heteromers were found able to control the release of glutamate from the striatal astrocyte processes. A2A-D2 heteromers on striatal astrocytes and astrocyte processes are discussed as far as their potential relevance in the control of glutamatergic transmission in striatum is concerned, including potential roles in glutamatergic transmission dysregulation in pathological conditions including schizophrenia or the Parkinson's disease.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Italy.
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Cristina Ceccoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy.
| | - Luigi F Agnati
- Department of Biochemical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; Center for Promotion of 3Rs in Teaching and Research (Centro 3R), Pisa, Italy; Center of Excellence for Biomedical Research, University of Genova, Italy.
| |
Collapse
|
8
|
Woitalla D, Buhmann C, Hilker-Roggendorf R, Höglinger G, Koschel J, Müller T, Weise D. Role of dopamine agonists in Parkinson's disease therapy. J Neural Transm (Vienna) 2023; 130:863-873. [PMID: 37165120 DOI: 10.1007/s00702-023-02647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/27/2023] [Indexed: 05/12/2023]
Abstract
Dopamine agonists are an important component of Parkinson's therapy. When weighing up the various therapy options, therapy with levodopa has recently been increasingly preferred due to its stronger efficacy and the ostensibly lower rate of side effects. The advantage of the lower incidence of motor complications during therapy with dopamine agonists was neglected. The occurrence of side effects can be explained by the different receptor affinity to the individual dopaminergic and non-dopaminergic receptors of the individual dopamine agonists. However, the different affinity to individual receptors also explains the different effect on individual Parkinson symptoms and can, therefore, contribute to a targeted use of the different dopamine agonists. Since comparative studies on the differential effect of dopamine agonists have only been conducted for individual substances, empirical knowledge of the differential effect is of great importance. Therefore, the guidelines for the treatment of Parkinson's disease do not consider the differential effect of the dopamine agonists. The historical consideration of dopamine agonists within Parkinson's therapy deserves special attention to be able to classify the current discussion about the significance of dopamine agonists.
Collapse
Affiliation(s)
- D Woitalla
- Department of Neurology, Katholische Kliniken Der Ruhrhalbinsel, Essen, Germany.
| | - C Buhmann
- Department of Neurology, Universitätsklinikum Hamburg, Hamburg, Germany
| | | | - G Höglinger
- Department of Neurology, Medizinische Hochschule Hannover, Hannover, Germany
| | - J Koschel
- Department of Neurology Parkinson-Klinik Ortenau, Wolfach, Germany
| | - T Müller
- Department of Neurology, Alexianer St. Joseph Krankenhaus, Berlin, Germany
| | - D Weise
- Department of Neurology, Asklepios Fachklinikum Stadtroda, Stadtroda, Germany
| |
Collapse
|
9
|
Lubec J, Hussein AM, Kalaba P, Feyissa DD, Arias-Sandoval E, Cybulska-Klosowicz A, Bezu M, Stojanovic T, Korz V, Malikovic J, Aher NY, Zehl M, Dragacevic V, Leban JJ, Sagheddu C, Wackerlig J, Pistis M, Correa M, Langer T, Urban E, Höger H, Lubec G. Low-Affinity/High-Selectivity Dopamine Transport Inhibition Sufficient to Rescue Cognitive Functions in the Aging Rat. Biomolecules 2023; 13:biom13030467. [PMID: 36979402 PMCID: PMC10046369 DOI: 10.3390/biom13030467] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
The worldwide increase in cognitive decline, both in aging and with psychiatric disorders, warrants a search for pharmacological treatment. Although dopaminergic treatment approaches represent a major step forward, current dopamine transporter (DAT) inhibitors are not sufficiently specific as they also target other transporters and receptors, thus showing unwanted side effects. Herein, we describe an enantiomerically pure, highly specific DAT inhibitor, S-CE-123, synthetized in our laboratory. Following binding studies to DAT, NET and SERT, GPCR and kinome screening, pharmacokinetics and a basic neurotoxic screen, S-CE-123 was tested for its potential to enhance and/or rescue cognitive functions in young and in aged rats in the non-invasive reward-motivated paradigm of a hole-board test for spatial learning. In addition, an open field study with young rats was carried out. We demonstrated that S-CE-123 is a low-affinity but highly selective dopamine reuptake inhibitor with good bioavailability. S-CE-123 did not induce hyperlocomotion or anxiogenic or stereotypic behaviour in young rats. Our compound improved the performance of aged but not young rats in a reward-motivated task. The well-described impairment of the dopaminergic system in aging may underlie the age-specific effect. We propose S-CE-123 as a possible candidate for developing a tentative therapeutic strategy for age-related cognitive decline and cognitive dysfunction in psychiatric disorders.
Collapse
Affiliation(s)
- Jana Lubec
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Ahmed M. Hussein
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Predrag Kalaba
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Daniel Daba Feyissa
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | | | - Anita Cybulska-Klosowicz
- Neurobiology of Emotions Laboratory, Nencki Institute of Experimental Biology, 02093 Warsaw, Poland
| | - Mekite Bezu
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Volker Korz
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Jovana Malikovic
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Nilima Y. Aher
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Martin Zehl
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Vladimir Dragacevic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Johann Jakob Leban
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Claudia Sagheddu
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy
| | - Judith Wackerlig
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Marco Pistis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, 09042 Monserrato, Italy
- Section of Cagliari, Neuroscience Institute, National Research Council of Italy (CNR), 09042 Cagliari, Italy
| | - Merce Correa
- Department of Psychobiology, Universitat Jaume I, 12006 Castelló, Spain
- Department of Psychological Sciences, Behavioral Neuroscience Division, University of Connecticut, Storrs, CT 06269, USA
| | - Thierry Langer
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Ernst Urban
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090 Vienna, Austria
| | - Harald Höger
- Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna, 2325 Himberg, Austria
| | - Gert Lubec
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-676-569-4816
| |
Collapse
|
10
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
11
|
Borroto-Escuela DO, Fuxe K. The integrative role of G protein-coupled receptor heterocomplexes in Parkinson's disease. Neural Regen Res 2022; 17:2211-2212. [PMID: 35259834 PMCID: PMC9083153 DOI: 10.4103/1673-5374.335792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
12
|
The mGlu 5 Receptor Protomer-Mediated Dopamine D 2 Receptor Trans-Inhibition Is Dependent on the Adenosine A 2A Receptor Protomer: Implications for Parkinson's Disease. Mol Neurobiol 2022; 59:5955-5969. [PMID: 35829830 PMCID: PMC9463353 DOI: 10.1007/s12035-022-02946-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 06/28/2022] [Indexed: 11/01/2022]
Abstract
The adenosine A2A receptor (A2AR), dopamine D2 receptor (D2R) and metabotropic glutamate receptor type 5 (mGluR5) form A2AR-D2R-mGluR5 heteroreceptor complexes in living cells and in rat striatal neurons. In the current study, we present experimental data supporting the view that the A2AR protomer plays a major role in the inhibitory modulation of the density and the allosteric receptor-receptor interaction within the D2R-mGluR5 heteromeric component of the A2AR-D2R-mGluR5 complex in vitro and in vivo. The A2AR and mGluR5 protomers interact and modulate D2R protomer recognition and signalling upon forming a trimeric complex from these receptors. Expression of A2AR in HEK293T cells co-expressing D2R and mGluR5 resulted in a significant and marked increase in the formation of the D2R-mGluR5 heteromeric component in both bioluminescence resonance energy transfer and proximity ligation assays. A highly significant increase of the the high-affinity component of D2R (D2RKi High) values was found upon cotreatment with the mGluR5 and A2AR agonists in the cells expressing A2AR, D2R and mGluR5 with a significant effect observed also with the mGluR5 agonist alone compared to cells expressing only D2R and mGluR5. In cells co-expressing A2AR, D2R and mGluR5, stimulation of the cells with an mGluR5 agonist like or D2R antagonist fully counteracted the D2R agonist-induced inhibition of the cAMP levels which was not true in cells only expressing mGluR5 and D2R. In agreement, the mGluR5-negative allosteric modulator raseglurant significantly reduced the haloperidol-induced catalepsy in mice, and in A2AR knockout mice, the haloperidol action had almost disappeared, supporting a functional role for mGluR5 and A2AR in enhancing D2R blockade resulting in catalepsy. The results represent a relevant example of integrative activity within higher-order heteroreceptor complexes.
Collapse
|
13
|
Guidolin D, Tortorella C, Marcoli M, Maura G, Agnati LF. Intercellular Communication in the Central Nervous System as Deduced by Chemical Neuroanatomy and Quantitative Analysis of Images: Impact on Neuropharmacology. Int J Mol Sci 2022; 23:5805. [PMID: 35628615 PMCID: PMC9145073 DOI: 10.3390/ijms23105805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023] Open
Abstract
In the last decades, new evidence on brain structure and function has been acquired by morphological investigations based on synergic interactions between biochemical anatomy approaches, new techniques in microscopy and brain imaging, and quantitative analysis of the obtained images. This effort produced an expanded view on brain architecture, illustrating the central nervous system as a huge network of cells and regions in which intercellular communication processes, involving not only neurons but also other cell populations, virtually determine all aspects of the integrative function performed by the system. The main features of these processes are described. They include the two basic modes of intercellular communication identified (i.e., wiring and volume transmission) and mechanisms modulating the intercellular signaling, such as cotransmission and allosteric receptor-receptor interactions. These features may also open new possibilities for the development of novel pharmacological approaches to address central nervous system diseases. This aspect, with a potential major impact on molecular medicine, will be also briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, 35121 Padova, Italy;
| | - Manuela Marcoli
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Guido Maura
- Department of Pharmacy, Center of Excellence for Biomedical Research, University of Genova, 16126 Genova, Italy; (M.M.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| |
Collapse
|
14
|
Abstract
Restless Legs Syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs that is often accompanied by periodic limb movements during sleep (PLMS). RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergics that target the D3 receptor. However, over time patients often develop a gradual tolerance that can lead to the emergence of adverse effects and the augmentation of the symptoms. While the basal ganglia and the striatum control leg movements, the lumbar spinal cord is the gateway for the sensory processing of the symptoms and critical for the associated leg movements. D3 receptors are highly expressed in nucleus accumbens (NAc) of the striatum and the sensory-processing areas of the spinal dorsal horn. In contrast, D1 receptors are strongly expressed throughout the entire striatum and in the ventral horn of the spinal cord. Long-term treatment with D3 receptor full agonists is associated with an upregulation of the D1 receptor subtype, and D3 and D1 receptors can form functional heteromers, in which the D3R controls the D1R function. It is conceivable that the switch from beneficial treatment to augmentation observed in RLS patients after prolonged D3R agonist exposure may be the result of unmasked D1-like receptor actions.
Collapse
Affiliation(s)
- Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
15
|
Fonseca-Barriendos D, Frías-Soria CL, Pérez-Pérez D, Gómez-López R, Borroto Escuela DO, Rocha L. Drug-resistant epilepsy: Drug target hypothesis and beyond the receptors. Epilepsia Open 2021; 7 Suppl 1:S23-S33. [PMID: 34542940 PMCID: PMC9340308 DOI: 10.1002/epi4.12539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Epilepsy is a chronic neurological disorder that affects more than 50 million people worldwide. Despite a recent introduction of antiseizure drugs for the treatment of epileptic seizures, one-third of these patients suffer from drug-resistant epilepsy (DRE). The therapeutic target hypothesis is a cited theory to explain DRE. According to the target hypothesis, the failure to achieve seizure freedom leads to alteration of the structure and/or function of the antiseizure medication (ASM) target. However, this hypothesis fails to explain why patients with DRE do not respond to antiseizure medications of different targets. This review presents different conditions, such as epigenetic mechanisms and protein-protein interactions that may result in alterations of diverse drug targets using different mechanisms. These novel conditions represent new targets to control DRE.
Collapse
Affiliation(s)
| | | | - Daniel Pérez-Pérez
- Plan of Combined Studies in Medicine (PECEM), Faculty of Medicine, UNAM, México City, Mexico
| | - Rosenda Gómez-López
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Mexico City, México
| | | | - Luisa Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, México City, México
| |
Collapse
|
16
|
Istradefylline Mitigates Age-Related Hearing Loss in C57BL/6J Mice. Int J Mol Sci 2021; 22:ijms22158000. [PMID: 34360766 PMCID: PMC8348536 DOI: 10.3390/ijms22158000] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/20/2022] Open
Abstract
Age-related hearing loss (ARHL) is the most common sensory disorder among older people, and yet, the treatment options are limited to medical devices such as hearing aids and cochlear implants. The high prevalence of ARHL mandates the development of treatment strategies that can prevent or rescue age-related cochlear degeneration. In this study, we investigated a novel pharmacological strategy based on inhibition of the adenosine A2A receptor (A2AR) in middle aged C57BL/6 mice prone to early onset ARHL. C57BL/6J mice were treated with weekly istradefylline (A2AR antagonist; 1 mg/kg) injections from 6 to 12 months of age. Auditory function was assessed using auditory brainstem responses (ABR) to tone pips (4–32 kHz). ABR thresholds and suprathreshold responses (wave I amplitudes and latencies) were evaluated at 6, 9, and 12 months of age. Functional outcomes were correlated with quantitative histological assessments of sensory hair cells. Cognitive function was assessed using the Morris water maze and the novel object recognition test, and the zero maze test was used to assess anxiety-like behaviour. Weekly injections of istradefylline attenuated ABR threshold shifts by approximately 20 dB at mid to high frequencies (16–32 kHz) but did not improve ABR suprathreshold responses. Istradefylline treatment improved hair cell survival in a turn-dependent manner, whilst the cognitive function was unaffected by istradefylline treatment. This study presents the first evidence for the rescue potential of istradefylline in ARHL and highlights the role of A2AR in development of age-related cochlear degeneration.
Collapse
|
17
|
Viaro R, Longo F, Vincenzi F, Varani K, Morari M. l-DOPA promotes striatal dopamine release through D1 receptors and reversal of dopamine transporter. Brain Res 2021; 1768:147583. [PMID: 34284020 DOI: 10.1016/j.brainres.2021.147583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 06/08/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022]
Abstract
Previous studies have pointed out that l-DOPA can interact with D1 or D2 receptors independent of its conversion to endogenous dopamine. The present study was set to investigate whether l-DOPA modulates dopamine release from striatal nerve terminals, using a preparation of synaptosomes preloaded with [3H]DA. Levodopa (1 µM) doubled the K+-induced [3H]DA release whereas the D2/D3 receptor agonist pramipexole (100 nM) inhibited it. The l-DOPA-evoked facilitation was mimicked by the D1 receptor agonist SKF38393 (30-300 nM) and prevented by the D1/D5 antagonist SCH23390 (100 nM) but not the DA transporter inhibitor GBR12783 (300 nM) or the aromatic l-amino acid decarboxylase inhibitor benserazide (1 µM). Higher l-DOPA concentrations (10 and 100 µM) elevated spontaneous [3H]DA efflux. This effect was counteracted by GBR12783 but not SCH23390. Binding of [3H]SCH23390 in synaptosomes (in test tubes) revealed a dense population of D1 receptors (2105 fmol/mg protein). Both SCH23390 and SKF38393 fully inhibited [3H]SCH23390 binding (Ki 0.42 nM and 29 nM, respectively). l-DOPA displaced [3H]SCH23390 binding maximally by 44% at 1 mM. This effect was halved by addition of GBR12935 and benserazide. We conclude that l-DOPA facilitates exocytotic [3H]DA release through SCH23390-sensitive D1 receptors, independent of its conversion to DA. It also promotes non-exocytotic [3H]DA release, possibly via conversion to DA and reversal of DA transporter. These data confirm that l-DOPA can directly interact with dopamine D1 receptors and might extend our knowledge of the neurobiological mechanisms underlying l-DOPA clinical effects.
Collapse
Affiliation(s)
- Riccardo Viaro
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy; Department of Neuroscience and Rehabilitation, Section of Physiology, University of Ferrara, via Fossato di Mortara 19, 44121 Ferrara, Italy
| | - Francesco Longo
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - Michele Morari
- Department of Neuroscience and Rehabilitation, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy.
| |
Collapse
|
18
|
Misganaw D. Heteromerization of dopaminergic receptors in the brain: Pharmacological implications. Pharmacol Res 2021; 170:105600. [PMID: 33836279 DOI: 10.1016/j.phrs.2021.105600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Dopamine exerts its physiological effects through two subtypes of receptors, i.e. the receptors of the D1 family (D1R and D5R) and the D2 family (D2R, D3R, and D4R), which differ in their pattern of distribution, affinity, and signaling. The D1-like subfamily (D1R and D5R) are coupled to Gαs/olf proteins to activate adenylyl cyclase whereas the D2-like receptors are coupled to Gαi/o subunits and suppress the activity of adenylyl cyclase. Dopamine receptors are capable of forming homodimers, heterodimers, and higher-order oligomeric complexes, resulting in a change in the individual protomers' recognition, signaling, and pharmacology. Heteromerization has the potential to modify the canonical pharmacological features of individual monomeric units such as ligand affinity, activation, signaling, and cellular trafficking through allosteric interactions, reviving the field and introducing a new pharmacological target. Since heteromers are expressed and formed in a tissue-specific manner, they could provide the framework to design selective and effective drug candidates, such as brain-penetrant heterobivalent drugs and interfering peptides, with limited side effects. Therefore, heteromerization could be a promising area of pharmacology research, as it could contribute to the development of novel pharmacological interventions for dopamine dysregulated brain disorders such as addiction, schizophrenia, cognition, Parkinson's disease, and other motor-related disorders. This review is articulated based on the three criteria established by the International Union of Basic and Clinical Pharmacology for GPCR heterodimers (IUPHAR): evidence of co-localization and physical interactions in native or primary tissue, presence of a new physiological and functional property than the individual protomers, and loss of interaction and functional fingerprints upon heterodimer disruption.
Collapse
Affiliation(s)
- Desye Misganaw
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Medicine and Health Science, Wollo University, P.O. Box 1145, Dessie, Ethiopia.
| |
Collapse
|
19
|
Borroto-Escuela DO, Wydra K, Fores-Pons R, Vasudevan L, Romero-Fernandez W, Frankowska M, Ferraro L, Beggiato S, Crespo-Ramirez M, Rivera A, Rocha LL, Perez de la Mora M, Stove C, Filip M, Fuxe K. The Balance of MU-Opioid, Dopamine D2 and Adenosine A2A Heteroreceptor Complexes in the Ventral Striatal-Pallidal GABA Antireward Neurons May Have a Significant Role in Morphine and Cocaine Use Disorders. Front Pharmacol 2021; 12:627032. [PMID: 33790790 PMCID: PMC8005530 DOI: 10.3389/fphar.2021.627032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022] Open
Abstract
The widespread distribution of heteroreceptor complexes with allosteric receptor-receptor interactions in the CNS represents a novel integrative molecular mechanism in the plasma membrane of neurons and glial cells. It was proposed that they form the molecular basis for learning and short-and long-term memories. This is also true for drug memories formed during the development of substance use disorders like morphine and cocaine use disorders. In cocaine use disorder it was found that irreversible A2AR-D2R complexes with an allosteric brake on D2R recognition and signaling are formed in increased densities in the ventral enkephalin positive striatal-pallidal GABA antireward neurons. In this perspective article we discuss and propose how an increase in opioid heteroreceptor complexes, containing MOR-DOR, MOR-MOR and MOR-D2R, and their balance with each other and A2AR-D2R complexes in the striatal-pallidal enkephalin positive GABA antireward neurons, may represent markers for development of morphine use disorders. We suggest that increased formation of MOR-DOR complexes takes place in the striatal-pallidal enkephalin positive GABA antireward neurons after chronic morphine treatment in part through recruitment of MOR from the MOR-D2R complexes due to the possibility that MOR upon morphine treatment can develop a higher affinity for DOR. As a result, increased numbers of D2R monomers/homomers in these neurons become free to interact with the A2A receptors found in high densities within such neurons. Increased numbers of A2AR-D2R heteroreceptor complexes are formed and contribute to enhanced firing of these antireward neurons due to loss of inhibitory D2R protomer signaling which finally leads to the development of morphine use disorder. Development of cocaine use disorder may instead be reduced through enkephalin induced activation of the MOR-DOR complex inhibiting the activity of the enkephalin positive GABA antireward neurons. Altogether, we propose that these altered complexes could be pharmacological targets to modulate the reward and the development of substance use disorders.
Collapse
Affiliation(s)
| | - Karolina Wydra
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Ramon Fores-Pons
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Lakshmi Vasudevan
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | | | - Małgorzata Frankowska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Sarah Beggiato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy
| | - Minerva Crespo-Ramirez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alicia Rivera
- Department of Cell Biology, University of Malaga, Instituto de Investigación Biomédica (IBIMA), Malaga, Spain
| | - Luisa L Rocha
- Pharmacobiology Department, Center for Research and Advanced Studies, Mexico City, Mexico
| | - Miguel Perez de la Mora
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Christophe Stove
- Laboratory of Toxicology, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
20
|
Martínez-Pinilla E, Rico AJ, Rivas-Santisteban R, Lillo J, Roda E, Navarro G, Lanciego JL, Franco R. Expression of GPR55 and either cannabinoid CB 1 or CB 2 heteroreceptor complexes in the caudate, putamen, and accumbens nuclei of control, parkinsonian, and dyskinetic non-human primates. Brain Struct Funct 2020; 225:2153-2164. [PMID: 32691218 DOI: 10.1007/s00429-020-02116-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
Endocannabinoids are neuromodulators acting on specific cannabinoid CB1 and CB2 G-protein-coupled receptors (GPCRs), representing potential therapeutic targets for neurodegenerative diseases. Cannabinoids also regulate the activity of GPR55, a recently "deorphanized" GPCR that directly interacts with CB1 and with CB2 receptors. Our hypothesis is that these heteromers may be taken as potential targets for Parkinson's disease (PD). This work aims at assessing the expression of heteromers made of GPR55 and CB1/CB2 receptors in the striatum of control and parkinsonian macaques (with and without levodopa-induced dyskinesia). For this purpose, double blind in situ proximity ligation assays, enabling the detection of GPCR heteromers in tissue samples, were performed in striatal sections of control, MPTP-treated and MPTP-treated animals rendered dyskinetic by chronic treatment with levodopa. Image analysis and statistical assessment were performed using dedicated software. We have previously demonstrated the formation of heteromers between GPR55 and CB1 receptor (CB1-GPR55_Hets), which is highly expressed in the central nervous system (CNS), but also with the CB2 receptor (CB2-GPR55_Hets). Compared to the baseline expression of CB1-GPR55_Hets in control animals, our results showed increased expression levels in basal ganglia input nuclei of MPTP-treated animals. These observed increases in CB1-GPR55_Hets returned back to baseline levels upon chronic treatment with levodopa in dyskinetic animals. Obtained data regarding CB2-GPR55_Hets were quite similar, with somehow equivalent amounts in control and dyskinetic animals, and with increased expression levels in MPTP animals. Taken together, the detected increased expression of GPR55-endocannabinoid heteromers appoints these GPCR complexes as potential non-dopaminergic targets for PD therapy.
Collapse
Affiliation(s)
- Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, Julián Clavería s/n, 33006, Asturias, Spain.
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Asturias, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain.
| | - Alberto J Rico
- Neurosciences Division, Centre for Applied Medical Research, CIMA, University of Navarra, Avenida Pío XII, 55, 31008, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rafael Rivas-Santisteban
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, School of Chemistry, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
| | - Jaume Lillo
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, School of Chemistry, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain
| | - Elvira Roda
- Neurosciences Division, Centre for Applied Medical Research, CIMA, University of Navarra, Avenida Pío XII, 55, 31008, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain
| | - José Luis Lanciego
- Neurosciences Division, Centre for Applied Medical Research, CIMA, University of Navarra, Avenida Pío XII, 55, 31008, Pamplona, Spain.
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Rafael Franco
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, School of Chemistry, Universitat de Barcelona, Diagonal 643, 08028, Barcelona, Spain.
| |
Collapse
|
21
|
Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol 2020; 16:509-524. [PMID: 32641760 DOI: 10.1038/s41581-020-0304-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
ATP and its ultimate degradation product adenosine are potent extracellular signalling molecules that elicit a variety of pathophysiological functions in the kidney through the activation of P2 and P1 purinergic receptors, respectively. Extracellular purines can modulate immune responses, balancing inflammatory processes and immunosuppression; indeed, alterations in extracellular nucleotide and adenosine signalling determine outcomes of inflammation and healing processes. The functional activities of ectonucleotidases such as CD39 and CD73, which hydrolyse pro-inflammatory ATP to generate immunosuppressive adenosine, are therefore pivotal in acute inflammation. Protracted inflammation may result in aberrant adenosinergic signalling, which serves to sustain inflammasome activation and worsen fibrotic reactions. Alterations in the expression of ectonucleotidases on various immune cells, such as regulatory T cells and macrophages, as well as components of the renal vasculature, control purinergic receptor-mediated effects on target tissues within the kidney. The role of CD39 as a rheostat that can have an impact on purinergic signalling in both acute and chronic inflammation is increasingly supported by the literature, as detailed in this Review. Better understanding of these purinergic processes and development of novel drugs targeting these pathways could lead to effective therapies for the management of acute and chronic kidney disease.
Collapse
|
22
|
Yang P, Perlmutter JS, Benzinger TLS, Morris JC, Xu J. Dopamine D3 receptor: A neglected participant in Parkinson Disease pathogenesis and treatment? Ageing Res Rev 2020; 57:100994. [PMID: 31765822 PMCID: PMC6939386 DOI: 10.1016/j.arr.2019.100994] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by motor and non-motor symptoms which relentlessly and progressively lead to substantial disability and economic burden. Pathologically, these symptoms follow the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) associated with abnormal α-synuclein (α-Syn) deposition as cytoplasmic inclusions called Lewy bodies in pigmented brainstem nuclei, and in dystrophic neurons in striatal and cortical regions (Lewy neurites). Pharmacotherapy for PD focuses on improving quality of life and primarily targets dopaminergic pathways. Dopamine acts through two families of receptors, dopamine D1-like and dopamine D2-like; dopamine D3 receptors (D3R) belong to dopamine D2 receptor (D2R) family. Although D3R's precise role in the pathophysiology and treatment of PD has not been determined, we present evidence suggesting an important role for D3R in the early development and occurrence of PD. Agonist activation of D3R increases dopamine concentration, decreases α-Syn accumulation, enhances secretion of brain derived neurotrophic factors (BDNF), ameliorates neuroinflammation, alleviates oxidative stress, promotes neurogenesis in the nigrostriatal pathway, interacts with D1R to reduce PD associated motor symptoms and ameliorates side effects of levodopa (L-DOPA) treatment. Furthermore, D3R mutations can predict PD age of onset and prognosis of PD treatment. The role of D3R in PD merits further research. This review elucidates the potential role of D3R in PD pathogenesis and therapy.
Collapse
Affiliation(s)
- Pengfei Yang
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Physical Therapy, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Occupational Therapy, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Jinbin Xu
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. Adenosine A 2A-dopamine D 2 receptor-receptor interaction in neurons and astrocytes: Evidence and perspectives. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 169:247-277. [PMID: 31952688 DOI: 10.1016/bs.pmbts.2019.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The discovery of receptor-receptor interactions in the early 1980s, together with a more accurate focusing of allosteric mechanisms in proteins, expanded the knowledge on the G protein-coupled receptor (GPCR)-mediated signaling processes. GPCRs were seen to operate not only as monomers, but also as quaternary structures shaped by allosteric interactions. These integrative mechanisms can change the function of the GPCRs involved, leading to a sophisticated dynamic of the receptor assembly in terms of modulation of recognition and signaling. In this context, the heterodimeric complex formed by the adenosine A2A and the dopamine D2 receptors likely represents a prototypical example. The pharmacological evidence obtained, together with the tissue distribution of the A2A-D2 heteromeric complexes, suggested they could represent a target for new therapeutic strategies addressing significant disorders of the central nervous system. The research findings and the perspectives they offer from the therapeutic standpoint are the focus of the here presented discussion.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, Padova, Italy.
| | - Manuela Marcoli
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, Padova, Italy
| | - Guido Maura
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Alves ACDB, Bristot VJDO, Limana MD, Speck AE, Barros LSD, Solano AF, Aguiar AS. Role of Adenosine A 2A Receptors in the Central Fatigue of Neurodegenerative Diseases. J Caffeine Adenosine Res 2019. [DOI: 10.1089/caff.2019.0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Ana Cristina de Bem Alves
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | | | - Mirieli Denardi Limana
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Ana Elisa Speck
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| | - Leonardo Soares de Barros
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Alexandre Francisco Solano
- LABOX—Laboratório de Bioenergética e Estresse Oxidativo, Departamento de Bioquímica, UFSC—Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Aderbal S. Aguiar
- Exercise Biology Lab, Department of Health Sciences, UFSC—Universidade Federal de Santa Catarina, Araranguá, Brazil
| |
Collapse
|
25
|
Tóth A, Antal Z, Bereczki D, Sperlágh B. Purinergic Signalling in Parkinson's Disease: A Multi-target System to Combat Neurodegeneration. Neurochem Res 2019; 44:2413-2422. [PMID: 31054067 PMCID: PMC6776560 DOI: 10.1007/s11064-019-02798-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by progressive loss of dopaminergic neurons that results in characteristic motor and non-motor symptoms. L-3,4 dihydroxyphenylalanine (L-DOPA) is the gold standard therapy for the treatment of PD. However, long-term use of L-DOPA leads to side effects such as dyskinesias and motor fluctuation. Since purines have neurotransmitter and co-transmitter properties, the function of the purinergic system has been thoroughly studied in the nervous system. Adenosine and adenosine 5'-triphosphate (ATP) are modulators of dopaminergic neurotransmission, neuroinflammatory processes, oxidative stress, excitotoxicity and cell death via purinergic receptor subtypes. Aberrant purinergic receptor signalling can be either the cause or the result of numerous pathological conditions, including neurodegenerative disorders. Many data confirm the involvement of purinergic signalling pathways in PD. Modulation of purinergic receptor subtypes, the activity of ectonucleotidases and ATP transporters could be beneficial in the treatment of PD. We give a brief summary of the background of purinergic signalling focusing on its roles in PD. Possible targets for pharmacological treatment are highlighted.
Collapse
Affiliation(s)
- Adrián Tóth
- Department of Neurology, Faculty of Medicine, Semmelweis University, Balassa u. 6., Budapest, 1083, Hungary
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Üllői út 26., Budapest, 1085, Hungary
| | - Zsófia Antal
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary
| | - Dániel Bereczki
- Department of Neurology, Faculty of Medicine, Semmelweis University, Balassa u. 6., Budapest, 1083, Hungary
| | - Beáta Sperlágh
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43., Budapest, 1083, Hungary.
| |
Collapse
|
26
|
Borroto-Escuela DO, Fuxe K. Oligomeric Receptor Complexes and Their Allosteric Receptor-Receptor Interactions in the Plasma Membrane Represent a New Biological Principle for Integration of Signals in the CNS. Front Mol Neurosci 2019; 12:230. [PMID: 31607863 PMCID: PMC6773811 DOI: 10.3389/fnmol.2019.00230] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) not only exist as monomers but also as homomers and heteromers in which allosteric receptor-receptor interactions take place, modulating the functions of the participating GPCR protomers. GPCRs can also form heteroreceptor complexes with ionotropic receptors and receptor tyrosine kinases modulating their function. Furthermore, adaptor proteins interact with receptor protomers and modulate their interactions. The state of the art is that the allosteric receptor-receptor interactions are reciprocal, highly dynamic and substantially alter the signaling, trafficking, recognition and pharmacology of the participating protomers. The pattern of changes appears to be unique for each heteromer and can favor antagonistic or facilitatory interactions or switch the G protein coupling from e.g., Gi/o to Gq or to beta-arrestin signaling. It lends a new dimension to molecular integration in the nervous system. Future direction should be aimed at determining the receptor interface involving building models of selected heterodimers. This will make design of interface-interfering peptides that specifically disrupt the heterodimer possible. This will help to determine the functional role of the allosteric receptor-receptor interactions as well as the integration of signals at the plasma membrane by the heteroreceptor complexes, vs. integration of the intracellular signaling pathways. Integration of signals also at the plasma membrane seems crucial in view of the hypothesis that learning and memory at a molecular level takes place by reorganization of homo and heteroreceptor complexes in the postsynaptic membrane. Homo and heteroreceptor complexes are in balance with each other, and their disbalance is linked to disease. Targeting heteroreceptor complexes represents a novel strategy for the treatment of brain disorders.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, Urbino, Italy
- Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Fuxe K, Borroto-Escuela DO. Understanding receptor heteromerization and its allosteric integration of signals. Neuropharmacology 2019; 152:1-3. [PMID: 31054939 DOI: 10.1016/j.neuropharm.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, via Ca' le Suore 2, I-61029, Urbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100, Yaguajay, Cuba.
| |
Collapse
|
28
|
D3 and D1 receptors: The Yin and Yang in the treatment of restless legs syndrome with dopaminergics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 84:79-100. [PMID: 31229178 DOI: 10.1016/bs.apha.2019.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dopaminergic treatments targeting the D3 receptor subtype to reduce the symptoms of RLS show substantial initial clinical benefits but fail to maintain their efficacy over time. Sensorimotor circuits in the spinal cord are the gateway for the sensory processing of the symptoms and critical for the associated leg movements that relieve the symptoms and the periodic limb movements that often develop during sleep. There is a high preponderance of the inhibitory D3 receptor in the sensory-processing areas of the spinal cord (dorsal horn), whereas the motor areas in the ventral horn more strongly express the excitatory D1 receptor subtype. D3 and D1 receptors can form functional heteromeric ensembles that influence each other. In the spinal cord, long-term treatment with D3 receptor agonists is associated with the upregulation of the D1 receptor subtype and block of D1 receptor function at this stage can restore the D3 receptor effect. Alternate scenarios for a role of dopamine involve a role for the D5 receptor in regulating motor excitability and for the D4 receptor subtype in controlling D3-like effects. A model emerges that proposes that the behavioral changes in RLS, while responsive to D3 receptor agonists, may be ultimately be the result of unmasked increased D1-like receptor activities.
Collapse
|
29
|
Pelassa S, Guidolin D, Venturini A, Averna M, Frumento G, Campanini L, Bernardi R, Cortelli P, Buonaura GC, Maura G, Agnati LF, Cervetto C, Marcoli M. A2A-D2 Heteromers on Striatal Astrocytes: Biochemical and Biophysical Evidence. Int J Mol Sci 2019; 20:ijms20102457. [PMID: 31109007 PMCID: PMC6566402 DOI: 10.3390/ijms20102457] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Our previous findings indicate that A2A and D2 receptors are co-expressed on adult rat striatal astrocytes and on the astrocyte processes, and that A2A-D2 receptor–receptor interaction can control the release of glutamate from the processes. Functional evidence suggests that the receptor–receptor interaction was based on heteromerization of native A2A and D2 receptors at the plasma membrane of striatal astrocyte processes. We here provide biochemical and biophysical evidence confirming that receptor–receptor interaction between A2A and D2 receptors at the astrocyte plasma membrane is based on A2A-D2 heteromerization. To our knowledge, this is the first direct demonstration of the ability of native A2A and D2 receptors to heteromerize on glial cells. As striatal astrocytes are recognized to be involved in Parkinson’s pathophysiology, the findings that adenosine A2A and dopamine D2 receptors can form A2A-D2 heteromers on the astrocytes in the striatum (and that these heteromers can play roles in the control of the striatal glutamatergic transmission) may shed light on the molecular mechanisms involved in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Simone Pelassa
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Via Gabelli 63, 35122 Padova, Italy.
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Monica Averna
- Department of Experimental Medicine, Section of Biochemistry, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy.
| | - Giulia Frumento
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Letizia Campanini
- Division of Experimental Oncology, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy.
| | - Rosa Bernardi
- Division of Experimental Oncology, San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy.
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum-University of Bologna, Via Altura 3, 40139 Bologna, Italy.
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, 40139 Bologna, Italy.
| | - Giovanna Calandra Buonaura
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum-University of Bologna, Via Altura 3, 40139 Bologna, Italy.
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura 3, 40139 Bologna, Italy.
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy.
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 171 65 Stockholm, Sweden.
| | - Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy.
- Centre of Excellence for Biomedical Research CEBR, University of Genova, Viale Benedetto XV, 5, 16132 Genova, Italy.
| |
Collapse
|
30
|
Borroto-Escuela DO, Fuxe K. Adenosine heteroreceptor complexes in the basal ganglia are implicated in Parkinson's disease and its treatment. J Neural Transm (Vienna) 2019; 126:455-471. [PMID: 30637481 PMCID: PMC6456481 DOI: 10.1007/s00702-019-01969-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/06/2019] [Indexed: 02/08/2023]
Abstract
The adenosine homo, iso and heteroreceptor complexes in the basal ganglia play a highly significant role in modulating the indirect and direct pathways and the striosomal projections to the nigro-striatal DA system. The major adenosine receptor complexes in the striato-pallidal GABA neurons can be the A2AR-D2R and A2AR-D2R-mGluR5 receptor complexes, in which A2AR protomers and mGluR5 protomers can allosterically interact to inhibit D2R protomer signaling. Through a reorganization of these heteroreceptor complexes upon chronic dopaminergic treatment a pathological and prolonged inhibition of D2R receptor protomer signaling can develop with motor inhibition and wearing off of the therapeutic effects of levodopa and dopamine receptor agonists. The direct pathway is enriched in D1R in and around glutamate synapses enhancing the ability of these GABA neurons to be activated and increase motor initiation. The brake on these GABA neurons is in this case exerted by A1R forming A1R-D1R heteroreceptor complexes in which they allosterically inhibit D1R signaling and thereby reduce motor initiation. Upon chronic levodopa treatment a reorganization of the D1R heteroreceptor complexes develops with the formation of putative A1R-D1R-D3 in addition to D1R-D3R complexes in which D3R enhances D1R protomer signaling and may make the A1R protomer brake less effective. Alpha-synuclein monomers-dimers are postulated to form complexes with A2AR homo and heteroprotomers in the plasma membrane enhancing alpha-synuclein aggregation and toxicity. The alpha-synuclein fibrils formed in the A2AR enriched dendritic spines of the striato-pallidal GABA neurons may reach the surrounding DA terminals via extracellular-vesicle-mediated volume transmission involving internalization of the vesicles and their cargo (alpha-synuclein fibrils) into the vulnerable DA terminals, enhancing their degeneration followed by retrograde flow of these fibrils in the DA axons to the vulnerable nigral DA nerve cells.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Biomedicum, B0851, Solnavägen 9, 17177 Stockholm, Sweden
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100 Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Biomedicum, B0851, Solnavägen 9, 17177 Stockholm, Sweden
| |
Collapse
|
31
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. Receptor-Receptor Interactions as a Widespread Phenomenon: Novel Targets for Drug Development? Front Endocrinol (Lausanne) 2019; 10:53. [PMID: 30833931 PMCID: PMC6387912 DOI: 10.3389/fendo.2019.00053] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
The discovery of receptor-receptor interactions (RRI) has expanded our understanding of the role that G protein-coupled receptors (GPCRs) play in intercellular communication. The finding that GPCRs can operate as receptor complexes, and not only as monomers, suggests that several different incoming signals could already be integrated at the plasma membrane level via direct allosteric interactions between the protomers that form the complex. Most research in this field has focused on neuronal populations and has led to the identification of a large number of RRI. However, RRI have been seen to occur not only in neurons but also in astrocytes and, outside the central nervous system, in cells of the cardiovascular and endocrine systems and in cancer cells. Furthermore, RRI involving the formation of macromolecular complexes are not limited to GPCRs, being also observed in other families of receptors. Thus, RRI appear as a widespread phenomenon and oligomerization as a common mechanism for receptor function and regulation. The discovery of these macromolecular assemblies may well have a major impact on pharmacology. Indeed, the formation of receptor complexes significantly broadens the spectrum of mechanisms available to receptors for recognition and signaling, which may be implemented through modulation of the binding sites of the adjacent protomers and of their signal transduction features. In this context, the possible appearance of novel allosteric sites in the receptor complex structure may be of particular relevance. Thus, the existence of RRI offers the possibility of new therapeutic approaches, and novel pharmacological strategies for disease treatment have already been proposed. Several challenges, however, remain. These include the accurate characterization of the role that the receptor complexes identified so far play in pathological conditions and the development of ligands specific to given receptor complexes, in order to efficiently exploit the pharmacological properties of these complexes.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, Padova, Italy
- *Correspondence: Diego Guidolin
| | - Manuela Marcoli
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | | | - Guido Maura
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy
| | - Luigi F. Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of Adenosine Receptors: The State of the Art. Physiol Rev 2018; 98:1591-1625. [PMID: 29848236 DOI: 10.1152/physrev.00049.2017] [Citation(s) in RCA: 518] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adenosine is a ubiquitous endogenous autacoid whose effects are triggered through the enrollment of four G protein-coupled receptors: A1, A2A, A2B, and A3. Due to the rapid generation of adenosine from cellular metabolism, and the widespread distribution of its receptor subtypes in almost all organs and tissues, this nucleoside induces a multitude of physiopathological effects, regulating central nervous, cardiovascular, peripheral, and immune systems. It is becoming clear that the expression patterns of adenosine receptors vary among cell types, lending weight to the idea that they may be both markers of pathologies and useful targets for novel drugs. This review offers an overview of current knowledge on adenosine receptors, including their characteristic structural features, molecular interactions and cellular functions, as well as their essential roles in pain, cancer, and neurodegenerative, inflammatory, and autoimmune diseases. Finally, we highlight the latest findings on molecules capable of targeting adenosine receptors and report which stage of drug development they have reached.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Gessi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Stefania Merighi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Fabrizio Vincenzi
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| | - Katia Varani
- Department of Medical Sciences, University of Ferrara , Ferrara , Italy
| |
Collapse
|
33
|
Comorbidities, treatment, and pathophysiology in restless legs syndrome. Lancet Neurol 2018; 17:994-1005. [PMID: 30244828 DOI: 10.1016/s1474-4422(18)30311-9] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/05/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022]
Abstract
Restless legs syndrome, also known as Willis-Ekbom disease, is a common neurological condition whose manifestation is affected by complex environmental and genetic interactions. Restless legs syndrome can occur on its own, mostly at a young age, or with comorbidities such as cardiovascular disease, diabetes, and arterial hypertension, making it a difficult condition to properly diagnose. However, the concept of restless legs syndrome as being two entities, primary or secondary to another condition, has been challenged with genetic data providing further insight into the pathophysiology of the condition. Although dopaminergic treatment was formerly the first-line therapy, prolonged use can result in a serious worsening of symptoms known as augmentation. Clinical studies on pregabalin, gabapentin enacarbil, oxycodone-naloxone, and iron preparations have provided new treatment options, but most patients still report inadequate long-term management of symptoms. Studies of the hypoxic pathway activation and iron deficiency have provided valuable information about the pathophysiology of restless legs syndrome that should now be translated into new, more effective treatments for restless legs syndrome.
Collapse
|
34
|
Borroto-Escuela DO, Rodriguez D, Romero-Fernandez W, Kapla J, Jaiteh M, Ranganathan A, Lazarova T, Fuxe K, Carlsson J. Mapping the Interface of a GPCR Dimer: A Structural Model of the A 2A Adenosine and D 2 Dopamine Receptor Heteromer. Front Pharmacol 2018; 9:829. [PMID: 30214407 PMCID: PMC6125358 DOI: 10.3389/fphar.2018.00829] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022] Open
Abstract
The A2A adenosine (A2AR) and D2 dopamine (D2R) receptors form oligomers in the cell membrane and allosteric interactions across the A2AR–D2R heteromer represent a target for development of drugs against central nervous system disorders. However, understanding of the molecular determinants of A2AR–D2R heteromerization and the allosteric antagonistic interactions between the receptor protomers is still limited. In this work, a structural model of the A2AR–D2R heterodimer was generated using a combined experimental and computational approach. Regions involved in the heteromer interface were modeled based on the effects of peptides derived from the transmembrane (TM) helices on A2AR–D2R receptor–receptor interactions in bioluminescence resonance energy transfer (BRET) and proximity ligation assays. Peptides corresponding to TM-IV and TM-V of the A2AR blocked heterodimer interactions and disrupted the allosteric effect of A2AR activation on D2R agonist binding. Protein–protein docking was used to construct a model of the A2AR–D2R heterodimer with a TM-IV/V interface, which was refined using molecular dynamics simulations. Mutations in the predicted interface reduced A2AR–D2R interactions in BRET experiments and altered the allosteric modulation. The heterodimer model provided insights into the structural basis of allosteric modulation and the technique developed to characterize the A2AR–D2R interface can be extended to study the many other G protein-coupled receptors that engage in heteroreceptor complexes.
Collapse
Affiliation(s)
| | - David Rodriguez
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Wilber Romero-Fernandez
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jon Kapla
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Mariama Jaiteh
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Anirudh Ranganathan
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Tzvetana Lazarova
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience, Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Cervetto C, Venturini A, Guidolin D, Maura G, Passalacqua M, Tacchetti C, Cortelli P, Genedani S, Candiani S, Ramoino P, Pelassa S, Marcoli M, Agnati LF. Homocysteine and A2A-D2 Receptor-Receptor Interaction at Striatal Astrocyte Processes. J Mol Neurosci 2018; 65:456-466. [PMID: 30030763 DOI: 10.1007/s12031-018-1120-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/11/2018] [Indexed: 01/03/2023]
Abstract
The interaction between adenosine A2A and dopamine D2 receptors in striatal neurons is a well-established phenomenon and has opened up new perspectives on the molecular mechanisms involved in Parkinson's disease. However, it has barely been investigated in astrocytes. Here, we show by immunofluorescence that both A2A and D2 receptors are expressed in adult rat striatal astrocytes in situ, and investigate on presence, function, and interactions of the receptors in the astrocyte processes-acutely prepared from the adult rat striatum-and on the effects of homocysteine on the A2A-D2 receptor-receptor interaction. We found that A2A and D2 receptors were co-expressed on vesicular glutamate transporter-1-positive astrocyte processes, and confirmed that A2A-D2 receptor-receptor interaction controlled glutamate release-assessed by measuring the [3H]D-aspartate release-from the processes. The complexity of A2A-D2 receptor-receptor interaction is suggested by the effect of intracellular homocysteine, which reduced D2-mediated inhibition of glutamate release (homocysteine allosteric action on D2), without interfering with the A2A-mediated antagonism of the D2 effect (maintained A2A-D2 interaction). Our findings indicate the crucial integrative role of A2A-D2 molecular circuits at the plasma membrane of striatal astrocyte processes. The fact that homocysteine reduced D2-mediated inhibition of glutamate release could provide new insights into striatal astrocyte-neuron intercellular communications. As striatal astrocytes are recognized to be involved in Parkinson's pathophysiology, these findings may shed light on the pathogenic mechanisms of the disease and contribute to the development of new drugs for its treatment.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padova, Padua, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, Section of Biochemistry, and Italian Institute of Biostructures and Biosystems, University of Genova, Genoa, Italy
| | - Carlo Tacchetti
- Experimental Imaging Center, Scientific Institute San Raffaele, Milan, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DIBINEM) Alma Mater Studiorum, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Susanna Genedani
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Paola Ramoino
- Department of Earth, Environmental and Life Sciences, University of Genova, Genoa, Italy
| | - Simone Pelassa
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148, Genoa, Italy. .,Centre of Excellence for Biomedical Research CEBR, University of Genova, Genoa, Italy.
| | - Luigi F Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
36
|
Borroto-Escuela DO, Perez De La Mora M, Manger P, Narváez M, Beggiato S, Crespo-Ramírez M, Navarro G, Wydra K, Díaz-Cabiale Z, Rivera A, Ferraro L, Tanganelli S, Filip M, Franco R, Fuxe K. Brain Dopamine Transmission in Health and Parkinson's Disease: Modulation of Synaptic Transmission and Plasticity Through Volume Transmission and Dopamine Heteroreceptors. Front Synaptic Neurosci 2018; 10:20. [PMID: 30042672 PMCID: PMC6048293 DOI: 10.3389/fnsyn.2018.00020] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023] Open
Abstract
This perspective article provides observations supporting the view that nigro-striatal dopamine neurons and meso-limbic dopamine neurons mainly communicate through short distance volume transmission in the um range with dopamine diffusing into extrasynaptic and synaptic regions of glutamate and GABA synapses. Based on this communication it is discussed how volume transmission modulates synaptic glutamate transmission onto the D1R modulated direct and D2R modulated indirect GABA pathways of the dorsal striatum. Each nigro-striatal dopamine neuron was first calculated to form large numbers of neostriatal DA nerve terminals and then found to give rise to dense axonal arborizations spread over the neostriatum, from which dopamine is released. These neurons can through DA volume transmission directly influence not only the striatal GABA projection neurons but all the striatal cell types in parallel. It includes the GABA nerve cells forming the island-/striosome GABA pathway to the nigral dopamine cells, the striatal cholinergic interneurons and the striatal GABA interneurons. The dopamine modulation of the different striatal nerve cell types involves the five dopamine receptor subtypes, D1R to D5R receptors, and their formation of multiple extrasynaptic and synaptic dopamine homo and heteroreceptor complexes. These features of the nigro-striatal dopamine neuron to modulate in parallel the activity of practically all the striatal nerve cell types in the dorsal striatum, through the dopamine receptor complexes allows us to understand its unique and crucial fine-tuning of movements, which is lost in Parkinson's disease. Integration of striatal dopamine signals with other transmitter systems in the striatum mainly takes place via the receptor-receptor interactions in dopamine heteroreceptor complexes. Such molecular events also participate in the integration of volume transmission and synaptic transmission. Dopamine modulation of the glutamate synapses on the dorsal striato-pallidal GABA pathway involves D2R heteroreceptor complexes such as D2R-NMDAR, A2AR-D2R, and NTSR1-D2R heteroreceptor complexes. The dopamine modulation of glutamate synapses on the striato-entopeduncular/nigral pathway takes place mainly via D1R heteroreceptor complexes such as D1R-NMDAR, A2R-D1R, and D1R-D3R heteroreceptor complexes. Dopamine modulation of the island/striosome compartment of the dorsal striatum projecting to the nigral dopamine cells involve D4R-MOR heteroreceptor complexes. All these receptor-receptor interactions have relevance for Parkinson's disease and its treatment.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Physiology, Department of Biomolecular Science, University of Urbino, Urbino, Italy
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Yaguajay, Cuba
| | - Miguel Perez De La Mora
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paul Manger
- Faculty of Health Sciences, School of Anatomical Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Manuel Narváez
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Sarah Beggiato
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Minerva Crespo-Ramírez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
| | - Karolina Wydra
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Zaida Díaz-Cabiale
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Alicia Rivera
- Department of Cell Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - Luca Ferraro
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sergio Tanganelli
- Department of Life Sciences and Biotechnology (SVEB), University of Ferrara, Ferrara, Italy
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
- CiberNed: Centro de Investigación en Red Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Vecchio EA, Baltos JA, Nguyen ATN, Christopoulos A, White PJ, May LT. New paradigms in adenosine receptor pharmacology: allostery, oligomerization and biased agonism. Br J Pharmacol 2018; 175:4036-4046. [PMID: 29679502 DOI: 10.1111/bph.14337] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022] Open
Abstract
Adenosine receptors are a family of GPCRs containing four subtypes (A1 , A2A , A2B and A3 receptors), all of which bind the ubiquitous nucleoside adenosine. These receptors play an important role in physiology and pathophysiology and therefore represent attractive drug targets for a range of conditions. The theoretical framework surrounding drug action at adenosine receptors now extends beyond the notion of prototypical agonism and antagonism to encompass more complex pharmacological concepts. New paradigms include allostery, in which ligands bind a topographically distinct receptor site from that of the endogenous agonist, homomeric or heteromeric interactions across receptor oligomers and biased agonism, that is, ligand-dependent differential intracellular signalling. This review provides a concise overview of allostery, oligomerization and biased agonism at adenosine receptors and outlines how these paradigms may enhance future drug discovery endeavours focussed on the development of novel therapeutic agents acting at adenosine receptors. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Elizabeth A Vecchio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Anh T N Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| | - Paul J White
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Lauren T May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
38
|
Neuronal adenosine A 2A receptor overexpression is neuroprotective towards 3-nitropropionic acid-induced striatal toxicity: a rat model of Huntington's disease. Purinergic Signal 2018; 14:235-243. [PMID: 29770921 DOI: 10.1007/s11302-018-9609-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/01/2018] [Indexed: 10/16/2022] Open
Abstract
The A2A adenosine receptor (A2AR) is widely distributed on different cellular types in the brain, where it exerts a broad spectrum of pathophysiological functions, and for which a role in different neurodegenerative diseases has been hypothesized or demonstrated. To investigate the role of neuronal A2ARs in neurodegeneration, we evaluated in vitro and in vivo the effect of the neurotoxin 3-nitropropionic acid (3-NP) in a transgenic rat strain overexpressing A2ARs under the control of the neural-specific enolase promoter (NSEA2A rats). We recorded extracellular field potentials (FP) in corticostriatal slice and found that the synaptotoxic effect of 3-NP was significantly reduced in NSEA2A rats compared with wild-type animals (WT). In addition, after exposing corticostriatal slices to 3-NP 10 mM for 2 h, we found that striatal cell viability was significantly higher in NSEA2A rats compared to control rats. These in vitro results were confirmed by in vivo experiments: daily treatment of female rats with 3-NP 10 mg/kg for 8 days induced a selective bilateral lesion in the striatum, which was significantly reduced in NSEA2A compared to WT rats. These results demonstrate that the overexpression of the A2AR selectively at the neuronal level reduced 3-NP-induced neurodegeneration, and suggest an important function of the neuronal A2AR in the modulation of neurodegeneration.
Collapse
|
39
|
Borroto-Escuela DO, Tarakanov AO, Brito I, Fuxe K. Glutamate heteroreceptor complexes in the brain. Pharmacol Rep 2018; 70:936-950. [PMID: 32002960 DOI: 10.1016/j.pharep.2018.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/26/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022]
Abstract
The existence of mGluR, NMDAR, AMPAR and putative KAR heteroreceptor complexes in synaptic and extrasynaptic regions of brain glutamate synapses represents a major integrative mechanism. Our aim in the current article is to analyze if the formation of the different types glutamate hetereceptor complexes involves the contribution of triplet amino acid homologies (protriplets) in a postulated receptor interface based on the triplet puzzle theory. Seven main sets (lists) of receptor pairs in databases were used containing various sets (lists) of human receptor heteromers and nonheteromers obtained from the available scientific publications including the publically available GPCR-hetnet database. Brain mGluR1-mGluR5 and mGluR2-mGluR4 isoreceptor complexes were demonstrated with a predominant extrasynaptic localization at a post- and prejunctional localization. The existence of putative mGluR4-mGluR7 heteroreceptor complexes in the basal ganglia is proposed. Metabotropic glutamate receptor subtypes also participated in the formation of a large number of heteroreceptor complexes like mGluR1-A1R, mGluR5-A2AR, mGluR5-D2R and D2R-A2AR-mGluR5, located in relation to glutamate synapses, especially in the basal ganglia. A putative mGluR1-GABAB1/2 heterocomplex may also exist. NMDAR heteroreceptor complexes were also demonstrated as a fundamental integrative mechanism in the glutamate synapse and its extrasynaptic membranes. It represented fundamental work on inter alia NMDAR-mGluR5, NMDAR-D1R and NMDAR-D2R heteroreceptor complexes involving both antagonistic and facilitatory allosteric receptor-receptor interactions. As to AMPA receptors, a heterocomplex was found for the interaction between IFNgR1 and the AMPAR mediated via the subunit GluA1 which may be of relevance for neuroinflammation. AMPAR-D2R heteroreceptor complexes were also demonstrated. Besides glutamate heteroreceptor complexes and their allosteric receptor-receptor interactions, a significant mechanism for the functional crosstalk can also be phosphorylation and/or reorganization of adapter proteins with dynamic binding to the two receptors modulating the allosteric receptor mechanism.
Collapse
Affiliation(s)
- Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, Urbino, Italy.,Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Alexander O Tarakanov
- St. Petersburg Institute for Informatics and Automation, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Ismel Brito
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
40
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. G protein-coupled receptor-receptor interactions give integrative dynamics to intercellular communication. Rev Neurosci 2018; 29:703-726. [DOI: 10.1515/revneuro-2017-0087] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/01/2018] [Indexed: 01/14/2023]
Abstract
Abstract
The proposal of receptor-receptor interactions (RRIs) in the early 1980s broadened the view on the role of G protein-coupled receptors (GPCR) in the dynamics of the intercellular communication. RRIs, indeed, allow GPCR to operate not only as monomers but also as receptor complexes, in which the integration of the incoming signals depends on the number, spatial arrangement, and order of activation of the protomers forming the complex. The main biochemical mechanisms controlling the functional interplay of GPCR in the receptor complexes are direct allosteric interactions between protomer domains. The formation of these macromolecular assemblies has several physiologic implications in terms of the modulation of the signaling pathways and interaction with other membrane proteins. It also impacts on the emerging field of connectomics, as it contributes to set and tune the synaptic strength. Furthermore, recent evidence suggests that the transfer of GPCR and GPCR complexes between cells via the exosome pathway could enable the target cells to recognize/decode transmitters and/or modulators for which they did not express the pertinent receptors. Thus, this process may also open the possibility of a new type of redeployment of neural circuits. The fundamental aspects of GPCR complex formation and function are the focus of the present review article.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience , University of Padova, via Gabelli 65 , I-35121 Padova , Italy
| | - Manuela Marcoli
- Department of Pharmacy and Center of Excellence for Biomedical Research , University of Genova , I-16126 Genova , Italy
| | - Cinzia Tortorella
- Department of Neuroscience , University of Padova, via Gabelli 65 , I-35121 Padova , Italy
| | - Guido Maura
- Department of Pharmacy and Center of Excellence for Biomedical Research , University of Genova , I-16126 Genova , Italy
| | - Luigi F. Agnati
- Department of Biomedical Sciences , University of Modena and Reggio Emilia , I-41121 Modena , Italy
- Department of Neuroscience , Karolinska Institutet , S-17177 Stockholm , Sweden
| |
Collapse
|
41
|
Solís O, Moratalla R. Dopamine receptors: homomeric and heteromeric complexes in l-DOPA-induced dyskinesia. J Neural Transm (Vienna) 2018; 125:1187-1194. [DOI: 10.1007/s00702-018-1852-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
|
42
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
43
|
Borroto-Escuela DO, Narvaez M, Valladolid-Acebes I, Shumilov K, Di Palma M, Wydra K, Schaefer T, Reyes-Resina I, Navarro G, Mudó G, Filip M, Sartini S, Friedland K, Schellekens H, Beggiato S, Ferraro L, Tanganelli S, Franco R, Belluardo N, Ambrogini P, Pérez de la Mora M, Fuxe K. Detection, Analysis, and Quantification of GPCR Homo- and Heteroreceptor Complexes in Specific Neuronal Cell Populations Using the In Situ Proximity Ligation Assay. RECEPTOR-RECEPTOR INTERACTIONS IN THE CENTRAL NERVOUS SYSTEM 2018. [DOI: 10.1007/978-1-4939-8576-0_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Blesa J, Trigo-Damas I, Dileone M, Del Rey NLG, Hernandez LF, Obeso JA. Compensatory mechanisms in Parkinson's disease: Circuits adaptations and role in disease modification. Exp Neurol 2017; 298:148-161. [PMID: 28987461 DOI: 10.1016/j.expneurol.2017.10.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
The motor features of Parkinson's disease (PD) are well known to manifest only when striatal dopaminergic deficit reaches 60-70%. Thus, PD has a long pre-symptomatic and pre-motor evolution during which compensatory mechanisms take place to delay the clinical onset of disabling manifestations. Classic compensatory mechanisms have been attributed to changes and adjustments in the nigro-striatal system, such as increased neuronal activity in the substantia nigra pars compacta and enhanced dopamine synthesis and release in the striatum. However, it is not so clear currently that such changes occur early enough to account for the pre-symptomatic period. Other possible mechanisms relate to changes in basal ganglia and motor cortical circuits including the cerebellum. However, data from early PD patients are difficult to obtain as most studies have been carried out once the diagnosis and treatments have been established. Likewise, putative compensatory mechanisms taking place throughout disease evolution are nearly impossible to distinguish by themselves. Here, we review the evidence for the role of the best known and other possible compensatory mechanisms in PD. We also discuss the possibility that, although beneficial in practical terms, compensation could also play a deleterious role in disease progression.
Collapse
Affiliation(s)
- Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Inés Trigo-Damas
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Michele Dileone
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Natalia Lopez-Gonzalez Del Rey
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ledia F Hernandez
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
45
|
Fong R, Khokhar S, Chowdhury AN, Xie KG, Wong JHY, Fox AP, Xie Z. Caffeine accelerates recovery from general anesthesia via multiple pathways. J Neurophysiol 2017; 118:1591-1597. [PMID: 28659466 PMCID: PMC5596131 DOI: 10.1152/jn.00393.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/22/2017] [Accepted: 06/22/2017] [Indexed: 12/19/2022] Open
Abstract
Various studies have explored different ways to speed emergence from anesthesia. Previously, we have shown that three drugs that elevate intracellular cAMP (forskolin, theophylline, and caffeine) accelerate emergence from anesthesia in rats. However, our earlier studies left two main questions unanswered. First, were cAMP-elevating drugs effective at all anesthetic concentrations? Second, given that caffeine was the most effective of the drugs tested, why was caffeine more effective than forskolin since both drugs elevate cAMP? In our current study, emergence time from anesthesia was measured in adult rats exposed to 3% isoflurane for 60 min. Caffeine dramatically accelerated emergence from anesthesia, even at the high level of anesthetic employed. Caffeine has multiple actions including blockade of adenosine receptors. We show that the selective A2a adenosine receptor antagonist preladenant or the intracellular cAMP ([cAMP]i)-elevating drug forskolin, accelerated recovery from anesthesia. When preladenant and forskolin were tested together, the effect on anesthesia recovery time was additive indicating that these drugs operate via different pathways. Furthermore, the combination of preladenant and forskolin was about as effective as caffeine suggesting that both A2A receptor blockade and [cAMP]i elevation play a role in caffeine's ability to accelerate emergence from anesthesia. Because anesthesia in rodents is thought to be similar to that in humans, these results suggest that caffeine might allow for rapid and uniform emergence from general anesthesia in humans at all anesthetic concentrations and that both the elevation of [cAMP]i and adenosine receptor blockade play a role in this response.NEW & NOTEWORTHY Currently, there is no method to accelerate emergence from anesthesia. Patients "wake" when they clear the anesthetic from their systems. Previously, we have shown that caffeine can accelerate emergence from anesthesia. In this study, we show that caffeine is effective even at high levels of anesthetic. We also show that caffeine operates by both elevating intracellular cAMP levels and by blocking adenosine receptors. This complicated pharmacology makes caffeine especially effective in accelerating emergence from anesthesia.
Collapse
Affiliation(s)
- Robert Fong
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois
| | - Suhail Khokhar
- College of Medicine, University of Illinois, School of Life Sciences, Chicago, Illinois
| | - Atif N Chowdhury
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kelvin G Xie
- School of Engineering and Applied Science, Washington University, St Louis, Missouri
| | | | - Aaron P Fox
- Department of Neurobiology, Pharmacology and Physiology, The University of Chicago, Chicago, Illinois
| | - Zheng Xie
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, Illinois;
| |
Collapse
|
46
|
Sanjari Moghaddam H, Zare-Shahabadi A, Rahmani F, Rezaei N. Neurotransmission systems in Parkinson’s disease. Rev Neurosci 2017; 28:509-536. [DOI: 10.1515/revneuro-2016-0068] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/10/2017] [Indexed: 12/17/2022]
Abstract
AbstractParkinson’s disease (PD) is histologically characterized by the accumulation of α-synuclein particles, known as Lewy bodies. The second most common neurodegenerative disorder, PD is widely known because of the typical motor manifestations of active tremor, rigidity, and postural instability, while several prodromal non-motor symptoms including REM sleep behavior disorders, depression, autonomic disturbances, and cognitive decline are being more extensively recognized. Motor symptoms most commonly arise from synucleinopathy of nigrostriatal pathway. Glutamatergic, γ-aminobutyric acid (GABA)ergic, cholinergic, serotoninergic, and endocannabinoid neurotransmission systems are not spared from the global cerebral neurodegenerative assault. Wide intrabasal and extrabasal of the basal ganglia provide enough justification to evaluate network circuits disturbance of these neurotransmission systems in PD. In this comprehensive review, English literature in PubMed, Science direct, EMBASE, and Web of Science databases were perused. Characteristics of dopaminergic and non-dopaminergic systems, disturbance of these neurotransmitter systems in the pathophysiology of PD, and their treatment applications are discussed.
Collapse
Affiliation(s)
- Hossein Sanjari Moghaddam
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Student Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Zare-Shahabadi
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImmunology Research Association (NIRA), Universal Scientific Education and Research Network (USERN), Tehran 1419783151, Iran
- Psychiatry and Psychology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Rahmani
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1419783151, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Boston, MA, USA
| |
Collapse
|
47
|
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pathological overproduction: the bad side of adenosine. Br J Pharmacol 2017; 174:1945-1960. [PMID: 28252203 PMCID: PMC6398520 DOI: 10.1111/bph.13763] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an endogenous ubiquitous purine nucleoside, which is increased by hypoxia, ischaemia and tissue damage and mediates a number of physiopathological effects by interacting with four GPCRs, identified as A1 , A2A , A2B and A3 . Physiological and acutely increased adenosine is mostly associated with beneficial effects that include vasodilatation and a decrease in inflammation. In contrast, chronic overproduction of adenosine occurs in important pathological states, where long-lasting increases in the nucleoside levels are responsible for the bad side of adenosine associated with chronic inflammation, fibrosis and organ damage. In this review, we describe and critically discuss the pathological overproduction of adenosine and analyse when, where and how adenosine exerts its detrimental effects throughout the body.
Collapse
Affiliation(s)
- Pier Andrea Borea
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Gessi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Stefania Merighi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Fabrizio Vincenzi
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| | - Katia Varani
- Department of Medical SciencesUniversity of FerraraFerrara44121Italy
| |
Collapse
|
48
|
Regulation of G Protein-Coupled Receptors by Ubiquitination. Int J Mol Sci 2017; 18:ijms18050923. [PMID: 28448471 PMCID: PMC5454836 DOI: 10.3390/ijms18050923] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/20/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
Abstract
G protein-coupled receptors (GPCRs) comprise the largest family of membrane receptors that control many cellular processes and consequently often serve as drug targets. These receptors undergo a strict regulation by mechanisms such as internalization and desensitization, which are strongly influenced by posttranslational modifications. Ubiquitination is a posttranslational modification with a broad range of functions that is currently gaining increased appreciation as a regulator of GPCR activity. The role of ubiquitination in directing GPCRs for lysosomal degradation has already been well-established. Furthermore, this modification can also play a role in targeting membrane and endoplasmic reticulum-associated receptors to the proteasome. Most recently, ubiquitination was also shown to be involved in GPCR signaling. In this review, we present current knowledge on the molecular basis of GPCR regulation by ubiquitination, and highlight the importance of E3 ubiquitin ligases, deubiquitinating enzymes and β-arrestins. Finally, we discuss classical and newly-discovered functions of ubiquitination in controlling GPCR activity.
Collapse
|
49
|
Adenosine A1 and A2A Receptors in the Brain: Current Research and Their Role in Neurodegeneration. Molecules 2017; 22:molecules22040676. [PMID: 28441750 PMCID: PMC6154612 DOI: 10.3390/molecules22040676] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
The inhibitory adenosine A1 receptor (A1R) and excitatory A2A receptor (A2AR) are predominantly expressed in the brain. Whereas the A2AR has been implicated in normal aging and enhancing neurotoxicity in multiple neurodegenerative diseases, the inhibitory A1R has traditionally been ascribed to have a neuroprotective function in various brain insults. This review provides a summary of the emerging role of prolonged A1R signaling and its potential cross-talk with A2AR in the cellular basis for increased neurotoxicity in neurodegenerative disorders. This A1R signaling enhances A2AR-mediated neurodegeneration, and provides a platform for future development of neuroprotective agents in stroke, Parkinson’s disease and epilepsy.
Collapse
|
50
|
Borroto-Escuela DO, Carlsson J, Ambrogini P, Narváez M, Wydra K, Tarakanov AO, Li X, Millón C, Ferraro L, Cuppini R, Tanganelli S, Liu F, Filip M, Diaz-Cabiale Z, Fuxe K. Understanding the Role of GPCR Heteroreceptor Complexes in Modulating the Brain Networks in Health and Disease. Front Cell Neurosci 2017; 11:37. [PMID: 28270751 PMCID: PMC5318393 DOI: 10.3389/fncel.2017.00037] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022] Open
Abstract
The introduction of allosteric receptor-receptor interactions in G protein-coupled receptor (GPCR) heteroreceptor complexes of the central nervous system (CNS) gave a new dimension to brain integration and neuropsychopharmacology. The molecular basis of learning and memory was proposed to be based on the reorganization of the homo- and heteroreceptor complexes in the postjunctional membrane of synapses. Long-term memory may be created by the transformation of parts of the heteroreceptor complexes into unique transcription factors which can lead to the formation of specific adapter proteins. The observation of the GPCR heterodimer network (GPCR-HetNet) indicated that the allosteric receptor-receptor interactions dramatically increase GPCR diversity and biased recognition and signaling leading to enhanced specificity in signaling. Dysfunction of the GPCR heteroreceptor complexes can lead to brain disease. The findings of serotonin (5-HT) hetero and isoreceptor complexes in the brain over the last decade give new targets for drug development in major depression. Neuromodulation of neuronal networks in depression via 5-HT, galanin peptides and zinc involve a number of GPCR heteroreceptor complexes in the raphe-hippocampal system: GalR1-5-HT1A, GalR1-5-HT1A-GPR39, GalR1-GalR2, and putative GalR1-GalR2-5-HT1A heteroreceptor complexes. The 5-HT1A receptor protomer remains a receptor enhancing antidepressant actions through its participation in hetero- and homoreceptor complexes listed above in balance with each other. In depression, neuromodulation of neuronal networks in the raphe-hippocampal system and the cortical regions via 5-HT and fibroblast growth factor 2 involves either FGFR1-5-HT1A heteroreceptor complexes or the 5-HT isoreceptor complexes such as 5-HT1A-5-HT7 and 5-HT1A-5-HT2A. Neuromodulation of neuronal networks in cocaine use disorder via dopamine (DA) and adenosine signals involve A2AR-D2R and A2AR-D2R-Sigma1R heteroreceptor complexes in the dorsal and ventral striatum. The excitatory modulation by A2AR agonists of the ventral striato-pallidal GABA anti-reward system via targeting the A2AR-D2R and A2AR-D2R-Sigma1R heteroreceptor complex holds high promise as a new way to treat cocaine use disorders. Neuromodulation of neuronal networks in schizophrenia via DA, adenosine, glutamate, 5-HT and neurotensin peptides and oxytocin, involving A2AR-D2R, D2R-NMDAR, A2AR-D2R-mGluR5, D2R-5-HT2A and D2R-oxytocinR heteroreceptor complexes opens up a new world of D2R protomer targets in the listed heterocomplexes for treatment of positive, negative and cognitive symptoms of schizophrenia.
Collapse
Affiliation(s)
- Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska InstitutetStockholm, Sweden; Department of Biomolecular Science, Section of Physiology, University of UrbinoUrbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-EstudioYaguajay, Cuba
| | - Jens Carlsson
- Department of Cell and Molecular Biology, Uppsala Biomedical Centre (BMC), Uppsala University Uppsala, Sweden
| | - Patricia Ambrogini
- Department of Biomolecular Science, Section of Physiology, University of Urbino Urbino, Italy
| | - Manuel Narváez
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga Málaga, Spain
| | - Karolina Wydra
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences Kraków, Poland
| | - Alexander O Tarakanov
- St. Petersburg Institute for Informatics and Automation, Russian Academy of Sciences Saint Petersburg, Russia
| | - Xiang Li
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| | - Carmelo Millón
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga Málaga, Spain
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara Ferrara, Italy
| | - Riccardo Cuppini
- Department of Biomolecular Science, Section of Physiology, University of Urbino Urbino, Italy
| | - Sergio Tanganelli
- Department of Medical Sciences, University of Ferrara Ferrara, Italy
| | - Fang Liu
- Campbell Research Institute, Centre for Addiction and Mental Health, University of Toronto Toronto, ON, Canada
| | - Malgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences Kraków, Poland
| | - Zaida Diaz-Cabiale
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga Málaga, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|