Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stefano Espinoza is active.

Publication


Featured researches published by Stefano Espinoza.


British Journal of Pharmacology | 2015

Dopamine receptors – IUPHAR Review 13

Jean-Martin Beaulieu; Stefano Espinoza; Raul R. Gainetdinov

The variety of physiological functions controlled by dopamine in the brain and periphery is mediated by the D1, D2, D3, D4 and D5 dopamine GPCRs. Drugs acting on dopamine receptors are significant tools for the management of several neuropsychiatric disorders including schizophrenia, bipolar disorder, depression and Parkinsons disease. Recent investigations of dopamine receptor signalling have shown that dopamine receptors, apart from their canonical action on cAMP‐mediated signalling, can regulate a myriad of cellular responses to fine‐tune the expression of dopamine‐associated behaviours and functions. Such signalling mechanisms may involve alternate G protein coupling or non‐G protein mechanisms involving ion channels, receptor tyrosine kinases or proteins such as β‐arrestins that are classically involved in GPCR desensitization. Another level of complexity is the growing appreciation of the physiological roles played by dopamine receptor heteromers. Applications of new in vivo techniques have significantly furthered the understanding of the physiological functions played by dopamine receptors. Here we provide an update of the current knowledge regarding the complex biology, signalling, physiology and pharmacology of dopamine receptors.


Molecular Pharmacology | 2011

Functional interaction between trace amine-associated receptor 1 and dopamine D2 receptor.

Stefano Espinoza; Ali Salahpour; Bernard Masri; Tatyana D. Sotnikova; Mirko Messa; Larry S. Barak; Marc G. Caron; Raul R. Gainetdinov

The ability of dopamine receptors to interact with other receptor subtypes may provide mechanisms for modulating dopamine-related functions and behaviors. In particular, there is evidence suggesting that the trace amine-associated receptor 1 (TAAR1) affects the dopaminergic system by regulating the firing rate of dopaminergic neurons or by altering dopamine D2 receptor (D2R) responsiveness to ligands. TAAR1 is a Gαs protein-coupled receptor that is activated by biogenic amines, “trace amines,” such as β-phenylethylamine (β-PEA) and tyramine that are normally found at low concentrations in the mammalian brain. In the present study, we investigated the biochemical mechanism of interaction between TAAR1 and D2R and the role this interaction plays in D2R-related signaling and behaviors. Using a bioluminescence resonance energy transfer biosensor for cAMP, we demonstrated that the D2R antagonists haloperidol, raclopride, and amisulpride were able to enhance selectively a TAAR1-mediated β-PEA increase of cAMP. Moreover, TAAR1 and D2R were able to form heterodimers when coexpressed in human embryonic kidney 293 cells, and this direct interaction was disrupted in the presence of haloperidol. In addition, in mice lacking TAAR1, haloperidol-induced striatal c-Fos expression and catalepsy were significantly reduced. Taken together, these data suggest that TAAR1 and D2R have functional and physical interactions that could be critical for the modulation of the dopaminergic system by TAAR1 in vivo.


Frontiers in Endocrinology | 2012

BRET biosensors to study GPCR biology, pharmacology, and signal transduction.

Ali Salahpour; Stefano Espinoza; Bernard Masri; Vincent M. Lam; Larry S. Barak; Raul R. Gainetdinov

Bioluminescence resonance energy transfer (BRET)-based biosensors have been extensively used over the last decade to study protein–protein interactions and intracellular signal transduction in living cells. In this review, we discuss the various BRET biosensors that have been developed to investigate biology, pharmacology, and signaling of G protein-coupled receptors (GPCRs). GPCRs form two distinct types of multiprotein signal transduction complexes based upon their inclusion of G proteins or β-arrestins that can be differentially affected by drugs that exhibit functional selectivity toward G protein or β-arrestin signaling. BRET has been especially adept at illuminating the dynamics of protein–protein interactions between receptors, G proteins, β-arrestins, and their many binding partners in living cells; as well as measuring the formation and accumulation of second messengers following receptor activation. Specifically, we discuss in detail the application of BRET to study dopamine and trace amine receptors signaling, presenting examples of an exchange protein activated by cAMP biosensor to measure cAMP, β-arrestin biosensors to determine β-arrestin recruitment to the receptor, and dopamine D2 receptor and trace amine-associated receptor 1 biosensors to investigate heterodimerization between them. As the biochemical spectrum of BRET biosensors expands, the number of signaling pathways that can be measured will concomitantly increase. This will be particularly useful for the evaluation of functional selectivity in which the real-time BRET capability to measure distinct signaling modalities will dramatically shorten the time to characterize new generation of biased drugs. These emerging approaches will further expand the growing application of BRET in the screening for novel pharmacologically active compounds.


PLOS ONE | 2010

The Dopamine Metabolite 3-Methoxytyramine Is a Neuromodulator

Tatyana D. Sotnikova; Jean-Martin Beaulieu; Stefano Espinoza; Bernard Masri; Xiaodong Zhang; Ali Salahpour; Larry S. Barak; Marc G. Caron; Raul R. Gainetdinov

Dopamine (3-hydroxytyramine) is a well-known catecholamine neurotransmitter involved in multiple physiological functions including movement control. Here we report that the major extracellular metabolite of dopamine, 3-methoxytyramine (3-MT), can induce behavioral effects in a dopamine-independent manner and these effects are partially mediated by the trace amine associated receptor 1 (TAAR1). Unbiased in vivo screening of putative trace amine receptor ligands for potential effects on the movement control revealed that 3-MT infused in the brain is able to induce a complex set of abnormal involuntary movements in mice acutely depleted of dopamine. In normal mice, the central administration of 3-MT caused a temporary mild hyperactivity with a concomitant set of abnormal movements. Furthermore, 3-MT induced significant ERK and CREB phosphorylation in the mouse striatum, signaling events generally related to PKA-mediated cAMP accumulation. In mice lacking TAAR1, both behavioral and signaling effects of 3-MT were partially attenuated, consistent with the ability of 3-MT to activate TAAR1 receptors and cause cAMP accumulation as well as ERK and CREB phosphorylation in cellular assays. Thus, 3-MT is not just an inactive metabolite of DA, but a novel neuromodulator that in certain situations may be involved in movement control. Further characterization of the physiological functions mediated by 3-MT may advance understanding of the pathophysiology and pharmacology of brain disorders involving abnormal dopaminergic transmission, such as Parkinsons disease, dyskinesia and schizophrenia.


Neuropsychopharmacology | 2015

TAAR1 Modulates Cortical Glutamate NMDA Receptor Function

Stefano Espinoza; Gabriele Lignani; Lucia Caffino; Silvia Maggi; Ilya Sukhanov; Damiana Leo; Liudmila Mus; Marco Emanuele; Giuseppe Ronzitti; Anja Harmeier; Lucian Medrihan; Tatyana D. Sotnikova; Evelina Chieregatti; Marius C. Hoener; Fabio Benfenati; Valter Tucci; Fabio Fumagalli; Raul R. Gainetdinov

Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor expressed in the mammalian brain and known to influence subcortical monoaminergic transmission. Monoamines, such as dopamine, also play an important role within the prefrontal cortex (PFC) circuitry, which is critically involved in high-o5rder cognitive processes. TAAR1-selective ligands have shown potential antipsychotic, antidepressant, and pro-cognitive effects in experimental animal models; however, it remains unclear whether TAAR1 can affect PFC-related processes and functions. In this study, we document a distinct pattern of expression of TAAR1 in the PFC, as well as altered subunit composition and deficient functionality of the glutamate N-methyl-D-aspartate (NMDA) receptors in the pyramidal neurons of layer V of PFC in mice lacking TAAR1. The dysregulated cortical glutamate transmission in TAAR1-KO mice was associated with aberrant behaviors in several tests, indicating a perseverative and impulsive phenotype of mutants. Conversely, pharmacological activation of TAAR1 with selective agonists reduced premature impulsive responses observed in the fixed-interval conditioning schedule in normal mice. Our study indicates that TAAR1 plays an important role in the modulation of NMDA receptor-mediated glutamate transmission in the PFC and related functions. Furthermore, these data suggest that the development of TAAR1-based drugs could provide a novel therapeutic approach for the treatment of disorders related to aberrant cortical functions.


Neuropharmacology | 2015

Postsynaptic D2 dopamine receptor supersensitivity in the striatum of mice lacking TAAR1

Stefano Espinoza; Valentina Ghisi; Marco Emanuele; Damiana Leo; Ilya Sukhanov; Tatiana D. Sotnikova; Evelina Chieregatti; Raul R. Gainetdinov

Trace Amine-Associated Receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) known to modulate dopaminergic system through several mechanisms. Mice lacking this receptor show a higher sensitivity to dopaminergic stimuli, such as amphetamine; however, it is not clear whether D1 or D2 dopamine receptors and which associated intracellular signaling events are involved in this modulation. In the striatum of TAAR1 knock out (TAAR1-KO mice) we found that D2, but not D1, dopamine receptors were over-expressed, both in terms of mRNA and protein levels. Moreover, the D2 dopamine receptor-related G protein-independent AKT/GSK3 signaling pathway was selectively activated, as indicated by the decrease of phosphorylation of AKT and GSK3β. The decrease in phospho-AKT levels, suggesting an increase in D2 dopamine receptor activity in basal conditions, was associated with an increase of AKT/PP2A complex, as revealed by co-immunoprecipitation experiments. Finally, we found that the locomotor activation induced by the D2 dopamine receptor agonist quinpirole, but not by the full D1 dopamine receptor agonist SKF-82958, was increased in TAAR1-KO mice. These data demonstrate pronounced supersensitivity of postsynaptic D2 dopamine receptors in the striatum of TAAR1-KO mice and indicate that a close interaction of TAAR1 and D2 dopamine receptors at the level of postsynaptic structures has important functional consequences.


Chemical Biology & Drug Design | 2013

Insights into the Structure and Pharmacology of the Human Trace Amine-Associated Receptor 1 (hTAAR1): Homology Modelling and Docking Studies

Elena Cichero; Stefano Espinoza; Raul R. Gainetdinov; Livio Brasili; Paola Fossa

Trace amine‐associated receptor 1 (TAAR1) is a G protein–coupled receptor that belongs to the family of TAAR receptors and responds to a class of compounds called trace amines, such as β‐phenylethylamine (β‐PEA) and 3‐iodothyronamine (T1AM). The receptor is known to have a very rich pharmacology and could be also activated by other classes of compounds, including adrenergic and serotonergic ligands. It is expected that targeting TAAR1 could provide a novel pharmacological approach to correct monoaminergic dysfunctions found in several brain disorders, such as schizophrenia, depression, attention deficit hyperactivity disorder and Parkinson’s disease. Only recently, the first selective TAAR1 agonist RO5166017 has been identified. To explore the molecular mechanisms of protein–agonist interaction and speed up the identification of new chemical entities acting on this biomolecular target, we derived a homology model for the hTAAR1. The putative protein‐binding site has been explored by comparing the hTAAR1 model with the β2‐adrenoreceptor binding site, available by X‐ray crystallization studies, and with the homology modelled 5HT1A receptor. The obtained results, in tandem with docking studies performed with RO5166017, β‐PEA and T1AM, provided an opportunity to reasonably identify the hTAAR1 key residues involved in ligand recognition and thus define important starting points to design new agonists.


Chemical Biology & Drug Design | 2014

Further Insights Into the Pharmacology of the Human Trace Amine-Associated Receptors: Discovery of Novel Ligands for TAAR1 by a Virtual Screening Approach

Elena Cichero; Stefano Espinoza; Silvia Franchini; Sara Guariento; Livio Brasili; Raul R. Gainetdinov; Paola Fossa

Trace Amine‐Associated Receptor 1 (TAAR1) is a G protein‐coupled receptor that is expressed in brain and periphery and responds to a class of compounds called trace amines, such as β‐phenylethylamine (β‐PEA), tyramine, tryptamine, octopamine. The receptor is known to have a very rich pharmacology and could be also activated by different classes of compounds, including dopaminergic, adrenergic and serotonergic ligands. It is expected that targeting hTAAR1 could provide a novel pharmacological approach for several human disorders, such as schizophrenia, depression, attention deficit hyperactivity disorder, Parkinsons disease and metabolic diseases. Only recently, a small number of selective hTAAR1 agonists (among which RO5166017 and T1AM) and antagonist (EPPTB), have been reported in literature. With the aim to identify new molecular entities able to act as ligands for this target, we used an homology model for the hTAAR1 and performed a virtual screening procedure on an in‐house database of compounds. A number of interesting molecules were selected and by testing them in an in vitro assay we found several agonists and one antagonist, with activities in the low micromolar range. These compounds could represent the starting point for the development of more potent and selective TAAR1 ligands.


Journal of Medicinal Chemistry | 2015

Design, Synthesis, and Evaluation of Thyronamine Analogues as Novel Potent Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists.

Grazia Chiellini; Giulia Nesi; Maria Digiacomo; Rossella Malvasi; Stefano Espinoza; Martina Sabatini; Sabina Frascarelli; Annunziatina Laurino; Elena Cichero; Marco Macchia; Raul R. Gainetdinov; Paola Fossa; Laura Raimondi; Riccardo Zucchi; Simona Rapposelli

Trace amine associated receptor 1 (TAAR1) is a G protein coupled receptor (GPCR) expressed in brain and periphery activated by a wide spectrum of agonists that include, but are not limited to, trace amines (TAs), amphetamine-like psychostimulants, and endogenous thyronamines such as thyronamine (T0AM) and 3-iodothyronamine (T1AM). Such polypharmacology has made it challenging to understand the role and the biology of TAAR1. In an effort to understand the molecular basis of TAAR1 activation, we rationally designed and synthesized a small family of thyronamine derivatives. Among them, compounds 2 and 3 appeared to be a good mimic of the parent endogenous thyronamine, T0AM and T1AM, respectively, both in vitro and in vivo. Thus, these compounds offer suitable tools for studying the physiological roles of mouse TAAR1 and could represent the starting point for the development of more potent and selective TAAR1 ligands.


European Journal of Pharmacology | 2015

In-vivo pharmacology of Trace-Amine Associated Receptor 1.

Vincent M. Lam; Stefano Espinoza; Andrey S. Gerasimov; Raul R. Gainetdinov; Ali Salahpour

Trace-amines (TAs) are endogenous amines that are implicated in several physiological processes including modulation of aminergic neurotransmission. These compounds exert their effect by activating a class of G protein-coupled receptors termed Trace-Amine Associated Receptors (TAARs), where TAAR1 is the only human receptor that has been shown to bind endogenous TAs. Most of the studies have focused on studying the role of TAAR1 on modulation of the dopamine transmission. These studies indicate that TAAR1 is a negative regulator of dopamine transmission making TAAR1 a novel target for neuropsychiatric disorders that arises from dopamine dysfunction such as schizophrenia. This review discusses the unique pharmacology of TAAR1 with the major focus on the physiological role of TAAR1 and its modulation of dopamine transmission.

Collaboration


Dive into the Stefano Espinoza's collaboration.

Top Co-Authors

Avatar

Raul R. Gainetdinov

Saint Petersburg State University

View shared research outputs
Top Co-Authors

Avatar

Damiana Leo

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tatyana D. Sotnikova

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilya Sukhanov

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liudmila Mus

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Tatiana D. Sotnikova

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge