Skip to main content

HYPOTHESIS AND THEORY article

Front. Physiol., 07 June 2018
Sec. Integrative Physiology
This article is part of the Research Topic Physiological and Pathological Responses to Hypoxia and High Altitude View all 36 articles

Receptor–Receptor Interactions of G Protein-Coupled Receptors in the Carotid Body: A Working Hypothesis

  • 1Department of Neuroscience, University of Padua, Padua, Italy
  • 2Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy

In the carotid body (CB), a wide series of neurotransmitters and neuromodulators have been identified. They are mainly produced and released by type I cells and act on many different ionotropic and metabotropic receptors located in afferent nerve fibers, type I and II cells. Most metabotropic receptors are G protein-coupled receptors (GPCRs). In other transfected or native cells, GPCRs have been demonstrated to establish physical receptor–receptor interactions (RRIs) with formation of homo/hetero-complexes (dimers or receptor mosaics) in a dynamic monomer/oligomer equilibrium. RRIs modulate ligand binding, signaling, and internalization of GPCR protomers and they are considered of relevance for physiology, pharmacology, and pathology of the nervous system. We hypothesize that RRI may also occur in the different structural elements of the CB (type I cells, type II cells, and afferent fibers), with potential implications in chemoreception, neuromodulation, and tissue plasticity. This ‘working hypothesis’ is supported by literature data reporting the contemporary expression, in type I cells, type II cells, or afferent terminals, of GPCRs which are able to physically interact with each other to form homo/hetero-complexes. Functional data about cross-talks in the CB between different neurotransmitters/neuromodulators also support the hypothesis. On the basis of the above findings, the most significant homo/hetero-complexes which could be postulated in the CB include receptors for dopamine, adenosine, ATP, opioids, histamine, serotonin, endothelin, galanin, GABA, cannabinoids, angiotensin, neurotensin, and melatonin. From a methodological point of view, future studies should demonstrate the colocalization in close proximity (less than 10 nm) of the above receptors, through biophysical (i.e., bioluminescence/fluorescence resonance energy transfer, protein-fragment complementation assay, total internal reflection fluorescence microscopy, fluorescence correlation spectroscopy and photoactivated localization microscopy, X-ray crystallography) or biochemical (co-immunoprecipitation, in situ proximity ligation assay) methods. Moreover, functional approaches will be able to show if ligand binding to one receptor produces changes in the biochemical characteristics (ligand recognition, decoding, and trafficking processes) of the other(s). Plasticity aspects would be also of interest, as development and environmental stimuli (chronic continuous or intermittent hypoxia) produce changes in the expression of certain receptors which could potentially invest the dynamic monomer/oligomer equilibrium of homo/hetero-complexes and the correlated functional implications.

Introduction

The carotid body (CB) is a small polymodal peripheral chemoreceptor standing out for its basic role in case of hypoxia, hypercapnia, acidosis, low glucose; in these circumstances, it promotes an adequate respiratory and cardiovascular response as well as other less studied stimuli (Atanasova and Lazarov, 2014; Ortega-Sáenz et al., 2015). Two cell types can be distinguished in the CB, the ‘neuron-like’ chemosensitive type I cells and the ‘glial-like’ supportive type II cells. Type I cells synthetize and release many different neurotransmitters and neuromodulators, such as dopamine, acetylcholine, serotonin, histamine, ATP, glutamate, GABA, and substance P. Neurotransmitters and neuromodulators released by type I cells in great measure exert their activity on the afferent endings of the carotid sinus nerve (Porzionato et al., 2018), conveying the stimulation through the glossopharyngeal nerve and petrosal ganglion. The main excitatory stimuli are considered acetylcholine and ATP, binding to the ionotropic nicotinic and P2X2/P2X3 receptors, respectively. However, autocrine/paracrine modulation of type I cells is also of particular importance and is mainly performed through a series of metabotropic G protein-coupled receptors (GPCRs), such as A2A, D1/2, H1/2/3, M1/2, 5-HT2A, and others (for comprehensive reviews on neurotransmitters in the CB, see Iturriaga and Alcayaga, 2004; Bairam and Carroll, 2005; Nurse, 2005, 2014). Conversely, metabotropic GPCRs are also present in afferent nerve endings and type II cells, where they play a role in neuromodulation of signaling between type I cells and afferent fibers (Tse et al., 2012; Nurse et al., 2018). Many authors have emphasized the absolute peculiarity of the CB, characterized by “a plethora of neurotransmitters and neuromodulators, as well as an even broader spectrum of their receptors” (Nurse, 2014). Apart from the above sensory innervation, the CB also shows sensory innervation from jugular and nodose ganglia, post-ganglionic sympathetic nerve fibers from the superior cervical ganglion, and preganglionic parasympathetic and sympathetic fibers reaching ganglion cells in the CB. Efferent parasympathetic and sympathetic innervation of the CB plays a pivotal role in modulation of blood flow (De Caro et al., 2013).

-1The CB also shows a high level of structural plasticity, undergoing structural and functional changes during development (e.g., Porzionato et al., 2008a,b; De Caro et al., 2013), aging (e.g., Di Giulio et al., 2012; Zara et al., 2013) and as a consequence of environmental stimuli, such as chronic continuous hypoxia, taking place during acclimation to high altitude (e.g., Wang and Bisgard, 2002). In response to chronic hypoxia, type II cells can act in a stem cell-like manner, differentiating into precursors of neural cells that originate mature glomus cells (Pardal et al., 2007; Platero-Luengo et al., 2014).

The GPCR family involves about 800 human receptors that are organized into five subfamilies, namely classes A (the largest group), B, C, frizzled, and adhesion (Foord, 2002; Guidolin et al., 2018). Considering the structure, GPCRs have a similar conformation showing a peculiar seven TM (7TM) α-helical region, an extracellular amino-terminal segment and an intracellular carboxy-terminal tail (Tuteja, 2009). While the domain that is localized in the extracellular portion allows the ligand(s) – receptor interaction, the TM region is subject to a conformational change when the binding with a ligand occurs; this change is then transmitted to the intracellular region of the GPCR responsible for activating the signaling cascade (Schonenbach et al., 2015; Stockert and Devi, 2015).

According to evidences from in vitro and in vivo studies, GPCR monomers can recognize/decode signals (Bayburt et al., 2007; Whorton et al., 2007; Kuszak et al., 2009; Lohse, 2010; Guidolin et al., 2018); in particular, an intrinsic plasticity characterizes GPCR monomers signaling, in fact GPCR activation can determine the onset of different signal transduction patterns, like the G proteins and/or arrestin pathways (Zidar et al., 2009; Guidolin et al., 2018). Furthermore, it is established that GPCRs can functionally interact by sharing signaling pathways or by mechanisms of transactivation (see Köse, 2017; Guidolin et al., 2018). In the 1980s, however, Agnati et al. (2005) provided evidence suggesting that GPCRs could also establish structural receptor–receptor interactions (RRIs; see Guidolin et al., 2015, 2018 for recent reviews), in which they can associate with themselves (homodimerization) or with other proteins of the same family (heterodimerization). This concept was later confirmed in transfected mammalian cell systems but also in many different native tissues (i.e., central nervous system; mammary gland; liver; cancer tissues) (see Ferré et al., 2014; Guidolin et al., 2015). It is a fact that class C GPCRs form constitutive homomers or heteromers (Kniazeff et al., 2011; Guidolin et al., 2018). An example is represented by the metabotropic receptor for gamma-aminobutyric acid b (GABAB), that, in the central nervous system, constitutes the major inhibitory neurotransmitter (Calver et al., 2000; Pontier et al., 2006). The receptor for GABAB, which is formed by the subunits GABAB1 and GABAB2, represents the typical example of constitutive heterodimer (Lohse, 2010). Only GABAB1 can bind GABA; however, GABAB2, even if alone is not functional, is required as GABAB1 as such is considered immature due to the presence of a carboxy-terminal ER retention motif (Galvez et al., 2001; Kamal and Jockers, 2011). Hence, after co-expression, the heterodimerization is the prerequisite to permit the masking of the retention domain and the subsequent translation of the active protein to the plasma-membrane (Terrillon and Bouvier, 2004). The oligomerization process in class A GPCR is a broadly debated topic of interest (see Franco et al., 2016; Guidolin et al., 2018). However, according to the consistent results obtained through multiple approaches, the possibility of class A GPCR complexes is strongly supported (Bouvier and Hébert, 2014; Guidolin et al., 2018). In this respect, studies highlighting the occurrence of a specific and dynamic monomer/oligomer equilibrium in class A homodimers and heterodimers are of particular interest (Kroeger et al., 2003; Palczewski, 2010; Kleinau et al., 2016; Farran, 2017). In fact, according to their half-lives (determined by the association and dissociation rates) class A GPCR dimers are demonstrated to be often transient (Gurevich and Gurevich, 2008). This may contribute in explaining the opposing views regarding the role of class A GPCR monomers versus oligomers (Guidolin et al., 2018).

The amount of data proving the existence of GPCR heteromers showed a huge increase similarly to both development and diffusion of biophysical techniques able to detect the spatial proximity of protein molecules (Guidolin et al., 2015). They involve resonance energy transfer methods including bioluminescence and fluorescence resonance energy transfer (BRET and FRET, respectively), fluorescence complementation, total internal fluorescence microscopy, fluorescence correlation spectroscopy, assays based on bivalent ligands and proximity ligation assays (PLAs).

The basic molecular mechanism leading to the formation of the receptor assemblies are allosteric interactions (Kenakin et al., 2010; Fuxe et al., 2012a). As recently outlined by Changeux and Christopoulos (2016), the cooperativity that emerges in the action of orthosteric and allosteric ligands of the GPCR forming the complex provides the cell decoding apparatus with sophisticated dynamics in terms of recognition and signaling (George et al., 2002; Guidolin et al., 2018). It has been shown, for instance, that GPCR heterodimerization exerts effects by altering or fine-tuning ligand binding, signaling, as well as internalization of GPCR protomers (Bulenger et al., 2005; Satake et al., 2013; Gomes et al., 2016; Guidolin et al., 2018). Angiotensin II receptor (AT1) – bradykinin receptor (B2) heterodimer (detected in smooth muscle, omental vessel, and platelets) provides an example: angiotensin II triggers inositol triphosphate (IP3) accumulation in a manner that is much more potent and effective than in the cells showing the expression of AT1 alone; conversely, IP3 accumulation sustained by bradykinin is slightly weaker in cells expressing the AT1–B2 heterodimer with respect to the cells expressing only B2 (Satake et al., 2013).

Many evidences highlight that dimers may be formed by different mechanisms (Guidolin et al., 2018). Receptor complexes can arise in plasma membrane under the action of hydrophobic forces (Gahbauer and Böckmann, 2016). Receptor assembly may also occur in the endoplasmic reticulum (ER) before trafficking to the plasma membrane. ER is responsible of the structural quality of the synthetized receptors. Thus, only correctly folded receptors are allowed to exit while degradation occur for unfolded or misfolded proteins (Terrillon and Bouvier, 2004). Finally, recent data showed that GPCRs can be safely transferred by microvesicles from a source to a target cell where they become capable of recognizing and decoding their signal (Guescini et al., 2012).

Since GPCRs are implicated in several human diseases and represent major pharmacological targets, GPCR oligomerization process, determining variations in the processes of agonist recognition, signaling, and trafficking of participating receptors via allosteric mechanisms (Fuxe et al., 2012a), is of potential great importance for physiology and pharmacology, and new therapeutic strategies considering GPCR attitude to underwent toward several levels of receptor organization are under study (Farran, 2017).

Therefore, in a scenario where GPCR proteins are demonstrated to be present as monomer, the assumption that they may multimerize represents an interesting aspect to investigate. The purpose of the present article is to highlight indirect evidence of this with regard to the CB. In this tissue, indeed, the presence of multimeric GPCRs has not yet been consistently investigated. Thus, the main focus of the paper will be on possible homo-/heterodimers or mosaic receptors (already confirmed in other cell types) that could be present in the CB (type I and II cells, carotid sinus nerve afferents), with possible functional implications. The ‘working hypothesis’ of this paper could represent an interesting field of investigation aiming to clarify some aspects of chemoreception, neuromodulation and plasticity in the CB.

Investigative Approaches to GPCR Complexes

As briefly discussed before, the term RRI emphasizes the occurrence of an interaction needing a direct physical contact between the involved receptor proteins responsible for the formation of multimeric assemblies of receptors (dimers or high-order oligomers) at the cell membrane level, acting as integrative input units of membrane-associated molecular circuits (Guidolin et al., 2018). Considerable efforts are currently being directed toward the identification of the most adequate strategy to discover such a supramolecular organization of GPCRs, but no single method is free from limitations giving rise to some discrepancy between various studies (Mackie, 2005; Lambert and Javitch, 2014). Thus, evidence obtained through multiple approaches with consistent results is needed to identify receptor complexes (Bouvier and Hébert, 2014).

In particular, an operational definition of RRI (Fuxe et al., 2010) can be exploited to devise experimental procedures fulfilling the following two measurable conditions:

1. The binding of a ligand to one receptor determines a detectable change in the biochemical features of another receptor, that includes ligand recognition, decoding, and trafficking processes;

2. Involved protomers have not only to be colocalized, but also in close proximity to each other (less than 10 nm), as indicated by specific techniques.

To explore the second point, biophysical and biochemical methods are presently available (see Kaczor and Selent, 2011 for a review).

Biophysical Strategies

Biophysical methods may be divided into three macro-groups that are fluorescence/luminescence-based techniques, specialized microscopic techniques and X-ray crystallography.

As far as fluorescence/luminescence-based techniques are concerned, bioluminescence resonance energy transfer (BRET) and fluorescence resonance energy transfer (FRET) were among the earliest proximity-based assays of this type that were used in studying GPCR heteromerization (Fernández-Dueñas et al., 2012). Both techniques exploit the transfer of energy from a donor to an acceptor in a non-radiative manner (as a result of dipole–dipole coupling). If the donor is a fluorescent molecule with emission characteristics suitable to excite the acceptor, when they are in close proximity (∼10 nm) the result will be light emission from the acceptor fluorophore enabling the visualization of the subcellular location of specific receptor complexes.

Protein-fragment complementation assays have also been used to analyze GPCR heteromerization in living cells (Gandia et al., 2008). In this approach, cells are transfected with one GPCR fused to a fragment of fluorophore and another GPCR fused to a complementary fragment. Reconstitution of the active fluorophore would occur only if the two receptors were in close proximity. It is noteworthy that a combination of this technique with BRET or FRET can be devised to demonstrate the presence of GPCR trimers or tetramers (Gandia et al., 2008). In this respect, a specialized microscopic technique, namely atomic force microscopy, has also been explored to demonstrate the existence of GPCR complexes (Agnati et al., 2010).

However, all the mentioned techniques show limitations in answering questions concerning the dynamic nature of receptor complexes. Thus, more recently, the details of the spatial and temporal organization of GPCR complexes have been addressed by using specialized microscopic techniques allowing the direct observation of the state and behavior of individual proteins in living cells. They include total internal reflection fluorescence microscopy (Hern et al., 2010; Guidolin et al., 2018), fluorescence correlation spectroscopy (Chen et al., 2003; Guidolin et al., 2018) and photoactivated localization microscopy (PALM; Jonas et al., 2016).

X-ray crystallography represents a further significant experimental approach in the field. During the last years, crystallization techniques have been object of interest (Grisshammer, 2017), determining important consequences for the analysis of GPCR complexes as well as an increase of the number of experimentally assessed structures (Guidolin et al., 2018).

Biochemical Strategies

Spatial and temporal co-expression of GPCRs in cells and tissues is a necessary condition for the formation of more complex systems. This aspect can be investigated following biochemical approaches. The abundance of a specific mRNA can be detected by Northern-blot analysis, in situ hybridization, RT-PCR or microarray analysis. Immunohistochemistry and co-localization analysis (Agnati et al., 2005) can be performed to detect GPCR co-expression and co-localization at protein level. The practical limit of this technique regards the necessity for high-quality antibodies also due to low GPCRs expression levels.

To identify possible complex formation, it is mandatory for biochemical studies extracting GPCRs from the membrane by use of detergent, although shielding their native conformation in detergent is not a trivial task (Park et al., 2004).

Radiation inactivation technique (or target size method) take advantage of gamma rays or high energy electrons to disrupt polypeptides in order to identify the molecular weight of singular components. It is based on the inverse relationship existing between the size of a macromolecule and its dose-dependent inactivation. Hence, the probability to destroy a molecule increases along with its mass: larger is a protein and lower is the energy required to destroy it (Rios et al., 2001). For what it concerns GPCRs, the main limitation of the method is that this variation may be related to interaction with other proteins instead of oligomerization.

To overcome this limitation, co-immunoprecipitation can be used (Skieterska et al., 2013). Interestingly, this method in combination with confocal microscopy and FRET was used to detect homooligomerization of the 5-HT2C receptors as well as the 5-HT4 homodimerization in combination with BRET (Kaczor and Selent, 2011).

More recently, evidence of heteroreceptor complexes in native tissues has been provided by in situ PLA (Trifilieff et al., 2011). This technique could be adequate to highlight evidences regarding the presence of GPCR heteromers in tissue, giving new insights about basic biological mechanisms, heteroreceptor levels as well as their location. Thus, it allows to study the localization and modulation of heteroreceptor complexes, as formalin fixed tissue is used.

Cross-linking experiments are a further promising approach. They involve the use of assays based on bivalent ligands (agonists, antagonists, heteromer-selective antibodies, cross-linking reagents) allowing a direct targeting of the receptor complex (Yekkirala et al., 2013) in cells and tissues.

Homo- or Hetero-Dimers Between A1 and/or A2A Receptors

Adenosine receptor A1 homodimers have been identified in brain cortex of pigs by radioligand binding experiments (Ciruela et al., 1995). The capability of A1 receptors to form homodimers has also been demonstrated in Chinese hamster ovary (CHO) cells transfected with human A1 receptors (Gracia et al., 2008) and in transfected human embryonic kidney (HEK-293T) cells expressing similar levels of A1 receptor as the native tissue (Gracia et al., 2013). In bovine cortex, the existence of homomers was also shown through PLA (Gracia et al., 2013).

The effects of homodimerization on the receptor function can be inferred from ligand binding assays. A1 receptor activation inhibits adenylate cyclase and decreases cAMP concentration. Caffeine is a non-selective adenosine receptor antagonist. In case of low caffeine concentrations (when caffeine only binds to one protomer of the empty homodimer), it determines an increase of the agonist affinity for the other protomer in the A1 receptor homodimer; whereas, at high concentrations (when caffeine highly saturates both protomers of the homodimer) caffeine behaves like an A1 antagonist with a reduction of the agonist binding to the receptors. Thus, caffeine modulates A1 agonist-induced cAMP decrease in a biphasic manner. Interestingly, this is a particular behavior for a classical adenosine receptor antagonist like caffeine and is a pharmacological behavior explanation-lacking without taking into account receptor dimers as a minimal quaternary structure. Moreover, this pharmacological ability can also give explanations about the biphasic effects exerted at low and high concentration of caffeine on locomotor activity (Gracia et al., 2013).

As it regards the CB, adenosine is a well-known neurotransmitter produced in response to acute hypoxia. It can be directly released from type I cells, through an equilibrative nucleoside transporter, or it can be indirectly produced through breakdown of released ATP by ecto-5′-nucleotidase (Conde et al., 2012). Adenosine may act pre- and post-synaptically in the modulation of type I and afferents function. It enhances the firing pattern in the carotid sinus afferents through A2A receptors (Conde et al., 2009a,b; Piskuric and Nurse, 2013). Some discrepancies between studies have been reported about A1 localization into the CB structures (Bairam et al., 2009). Rocher et al. (1999) described the presence of A1 in rabbit type I cells, through functional studies on cell cultures. Conversely, in the rat, post-synaptic localization has mainly been suggested as A1 receptors are significantly expressed in the cytoplasm of nodous and petrosal ganglia (Bairam and Carroll, 2005; Gauda et al., 2006) and immunostaining of whole CB did not permit to specifically localize immunoreaction in type I cells (Bae et al., 2005). Possible species-related differences have been suggested and further analyses also on human material would be necessary. However, to reassume, A1 homodimerization may be hypothesized in nerve fibers and probably in those species expressing A1 receptors in type I cells.

A2A homodimerization has also been assessed in the plasma membrane of transfected HEK-293T cells by BRET, FRET, time-resolved BRET and immunoblotting and biotinylating experiments. Evidences highlight that more of 90% of A2A receptors are present as homodimers. In intracellular areas, the presence of a certain amount of monomeric species has been highlighted, suggesting that that they assemble into dimers before reaching cell surface (Canals et al., 2004; Lukasiewicz et al., 2007).

In particular, Canals et al. (2004) demonstrated that A2A homodimers are the functional species at the cell surface. Even though A2A homodimerization is quite constitutive, it is further influenced by specific ligands: agonists (i.e., CGS 21680) and antagonists (i.e., SCH 58261; caffeine) increase and decrease it, respectively (Lukasiewicz et al., 2007).

Various authors confirmed the presence of A2A receptor in the rat type I cells through several different techniques (RT-PCR studies, in situ hybridization analysis and immunohistochemistry) (e.g., Gauda, 2000; Kobayashi et al., 2000). Bairam et al. (2009) also demonstrated by Western-blot analysis the tendency of A2A receptor to form homodimer in the CB similarly to the superior cervical ganglion and nucleus tractus solitarius. This is one of the few specific references about receptor dimerization in the CB.

However, there are not studies addressing the issue of how agonist/antagonists may modulate dimerization of adenosine receptors in the nerve fibers and/or type I cells or, conversely, how dimerization may play a role in their pharmacological actions. It must also be considered that environmental conditions (hypoxia, etc.) may modulate receptor expression as well as types of dimerization, potentially modulating pharmacological effects.

Apart from the above homodimers, the ability of A1 receptors to heterodimerize with A2A receptors was also demonstrated in transfected cells by Ferré et al. (2008). Heterodimerization of these two protomers was assessed in vitro and in vivo by Ciruela et al. (2006), who confirmed the A1/A2A heterodimer formation in rat striatal glutamatergic nerve terminals by immunogold detection and co-immunoprecipitation. Recently it has been shown that at the presynaptic membrane of cortico-thalamic glutamatergic terminals A1 receptors co-localizes and interacts with A2A receptors, forming functional receptor heterodimer in the striatum (Fernández-Dueñas et al., 2017).

The eventual demonstration of A1/A2A heterodimers in type I cells and/or nerve fibers would be of particular interest in terms of modulation of the glutamatergic transmission. In fact, heterodimerization between the adenosine receptors A1 and A2A, which are responsible for opposite signaling pathways (inhibitory and excitatory actions, respectively) (Stockwell et al., 2016), has been suggested to exert a function in fine-tuning modulation of striatal glutamatergic neurotransmission by adenosine. Since A1 receptor shows higher affinity for adenosine than A2A, but A1 agonist affinity decreases when A2A is activated, the possibility exists that glutamate release may be inhibited or stimulated by a switch mechanism depending on low and high concentrations of adenosine, respectively (Ciruela et al., 2006; Doyle et al., 2012).

Hetero-Dimers Between Adenosine (A1 and A2A) and Purinergic (P2Y1, P2Y2, P2Y12) Receptors

P2Y1 (Xu et al., 2005; Tse et al., 2012) and P2Y12 (Carroll et al., 2012) receptors have been demonstrated in type I cells, where they inhibit the hypoxia-induced rise in intracellular Ca2+. Conversely, in type II cells, P2Y2 receptors have been demonstrated (Xu et al., 2003), which produces a rise in intracellular Ca2+.

Purinergic receptors have also been reported to form homodimers and hetero-dimers with other purinergic and adenosine receptors (e.g., Schicker et al., 2009). As previously detailed, A2A receptors are expressed in type I cells (Gauda, 2000; Kobayashi et al., 2000) and A1 receptors have been reported in the type I cells of some species (rabbit) (Rocher et al., 1999), although not confirmed in others (rat) (Gauda et al., 2000, 2006). Thus, in type I cells, many RRIs can be hypothesized: P2Y1/P2Y1, P2Y1/P2Y12, P2Y12/P2Y12, P2Y1/A1, P2Y1/A2A, P2Y12/A1, P2Y12/A2A (e.g., Yoshioka et al., 2001; Nakata et al., 2005, 2010; Schicker et al., 2009). Physical interactions between A1 and P2Y1 receptors, for instance, produce a conformational change in the A1 binding pocket with acquisition of P2Y1-like agonistic pharmacology, i.e., a P2Y1 agonist may bind to the A1 receptor and produce an inhibition of adenylate cyclase which is prevented by A1 antagonist (Fuxe et al., 2008). All the above RRIs would be particularly intriguing as they would represent other ways of reciprocal modulation between purinergic and adenosine neurotransmission both in type I and II cells.

Homo- and Hetero-Dimers Between Dopamine Receptors (D1 and D2)

According to co-immunoprecipitation data gathered from both rat brain and cells showing co-expression of the D1 and D2 receptors, occurs the idea they may constitute the same heteromeric protein complex; moreover, according to immunohistochemistry studies, these receptors are convincingly co-expressed and co-localized within neurons of human and rat brain (Lee et al., 2004). For instance, a significant neuronal subpopulation in rat nucleus accumbens co-expresses D1 and D2 receptors, which can form a D1/D2 receptor complex (Hopf et al., 2003; Hasbi et al., 2018). Rashid et al. (2007) also provided evidences concerning their co-expression in human embryonic kidney cells. In parallel, Rashid et al. (2007) and Hasbi et al. (2018) demonstrated the ability of D1 and D2 receptors to oligomerize in vivo in rodent and monkey striata, respectively, through in situ PLA, in situ FRET and co-immunoprecipitation.

D1 and D2 monomers are coupled to Gs and Gi proteins, respectively, and they are usually considered to exert opposite effects at the cellular level (Hopf et al., 2003). Conversely, the heterodimer D1/D2 is coupled to Gq/11. In the nucleus accumbens, the activation of this heterodimer-specific pathway determines an increase of calcium/calmodulin-dependent protein kinase IIα in contrast with the effect produced by the activation of the Gs-D1 receptor (Rashid et al., 2007).

Numerous studies documented the presence of D1 receptor in the CB of various animals through Northern blot analysis, RT-PCR and pharmacological studies, although the precise location has not been verified (Almaraz et al., 1991; Bairam et al., 1998, 2003; Schlenker, 2008). Almaraz et al. (1991) suggested expression in blood vessels. Bairam et al. (1998) suggested that D1 receptor could be expressed in nerve terminals (sensitive and sympathetic). Conversely, D1 receptors were identified in hamster type I cells through immunohistochemistry (Schlenker and Schultz, 2011). D2 has been demonstrated in type I cells and nerve fibers of the rat CB by double immunofluorescence (Wakai et al., 2015) and its expression is higher than D1 (Bairam et al., 1998). The contemporary presence of D1 and D2 in type I cells and nerve fibers would support the hypothesis of heterodimerization in these structures. Exogenous dopamine may also inhibit Ca2+ responses in type II cells, supporting the presence of corresponding receptors (Leonard and Nurse, 2017; Leonard et al., 2018) and of possible dimerization.

Huey and Powell (2000) explored the consequences of chronic hypoxia on D2 receptor expression in the arterial chemoreflex pathway. As regards the CB, the authors observed that the regulation of D2-receptor mRNA expression is time-dependent. Briefly, an initial increase of D2 receptor mRNA levels was observed after 6 and 12 h, with a subsequent decrease (24, 48 h) and a final significant increase after 168 h of hypoxia. In a noteworthy and analog study, Bairam et al. (2003) considered the effects induced by hypoxia on the expression levels of both D1 and D2 receptor mRNAs. The authors highlighted that in 1-day-old rabbits hypoxia affects the expression of D2- or D1-receptors mRNA in a manner that is age-dependent. D2- and D1-receptors transcript levels increased and decreased after exposure to moderate (15% O2) and severe (8% O2) hypoxia, respectively. Conversely, D2- and D1-transcript levels decreased independently of hypoxia intensities in adult rabbits. Unlike D1 receptors, changes in D2 receptor mRNA levels were independent on exposure time. The D2 role in the CB has been suggested not to be key under normal conditions, but rather in situations of chronic hypoxia, where D1 and D2 receptors density tends to change as previously discussed (Bairam et al., 2003).

The above changes in the expression of the different dopamine receptors after hypoxia suggest the possibility of consequent changes in the amount of D1/D2 heterodimers. It is intriguing the idea that D1/D2 heterodimer formation may be under the influence of environmental stimuli, mainly hypoxia but possibly others, in the CB.

Hetero-Dimers Between Adenosine (A1 and A2A) and Dopamine (D1 and D2) Receptors

The co-localization of A1 and D2 receptors has been showed by immunofluorescence in rat cerebral cortex neurons (Gines et al., 2000). Moreover, the presence of A1/D1 complexes was detected in mouse fibroblast Ltk-cells, transfected with human A1, D1, and D2. While the formation of A1/D1 heteromers was confirmed by coimmunoprecipitation, A1 did not seem to form heterodimers with D2. A1 and D1 receptors, separately, also tend to form homodimers; however, the preferred form of dimerization for these two receptors still needs to be elucidated (Agnati et al., 2003). Additionally, A1/D1 dimerization was also confirmed by FRET in HEK-293T cells (Shen et al., 2013).

Due to the possible presence of both A1 and D1 receptors in type I cells, a role by A1/D1 dimerization may be also be supposed. In particular, A1/D1 dimerization could represent a way of integration between agonists and/or antagonists for the two different monomers. For instance, Gines et al. (2000) suggested that A1/D1 heteromerization may have a role in the antagonistic modulation of D1 receptor in the brain, thus having an impact on desensitization mechanism of D1 and receptor trafficking.

A2A/D2 heterodimers have also been demonstrated both in transfected SH-SY5Y (Hillion et al., 2002; Xie et al., 2010) and HEK-293T cells (Navarro et al., 2014), by immunoprecipitation followed by Western-blotting (SH-SY5Y) and BRET (HEK-293T). In addition, A2A/D2 heterodimers were identified in neuronal primary cultures of rat striatum by PLA (Navarro et al., 2014) and cAMP accumulation experiments (Hillion et al., 2002). A2A/D2 heterodimers have also been demonstrated in the mammalian striatum, with particular reference to the striatal enkephalin-containing GABAergic neurons that project to the globus pallidus and comprise the so-called indirect pathway (Fink et al., 1992; Fuxe et al., 1998; Schiffmann et al., 2003; Trifilieff et al., 2011; Doyle et al., 2012; Atack et al., 2014). Thus, A2A/D2 heterodimers play an important role in the modulation of GABAergic striato-pallidal neuronal function (Bonaventura et al., 2015). In particular, antagonistic A2A-D2 RRIs occur in the heterodimer, as demonstrated in striatal membrane preparations after incubation with the A2A agonist CGS21680, leading to a decrease of the affinity of the high affinity D2 agonist-binding site (Fuxe et al., 1998; Guidolin et al., 2018). Relationships between A2A and D2 and adenosine/dopamine cross-talks have been proposed as possible new therapeutic approaches for Parkinson’s disease, schizophrenia, and drug addiction (Canals et al., 2003; Guidolin et al., 2015).

As previously discussed, both A2A and D2 are expressed in type I cells of the CB, supporting the hypothesis of A2A/D2 heterodimer formation also in these cells. The reciprocal influences of the two receptor monomers in the A2A/D2 complex would be particularly intriguing, as adenosine and dopamine are among the main excitatory and inhibitory neurotransmitters in the CB. Some authors have proposed a functional interaction between A2B and D2 receptors. In particular, if adenosine levels would become too high, activation of the low affinity A2B would lead to increased dopamine secretion from type I cells (Conde et al., 2006; Livermore and Nurse, 2013) and inhibition of sensory discharge through pre- and post-synaptic D2 receptors (Conde et al., 2009a,b, 2012; Zhang et al., 2017; Nurse et al., 2018). RRI between A2A and D2 could represent an opposite way of neuromodulation, increasing signaling efficiency in the presence of low adenosine levels, through allosteric receptor–receptor inhibition of D2 signaling.

In rat type I cells, Conde et al. (2008, 2009b) also proposed a A2B-D2 receptor interactions on the basis of in vitro pharmacological experiments, although they did not demonstrate if physical RRI occur but only demonstrated an interaction at the adenylyl cyclase level. In particular, they showed that D2 agonists decrease catecholamine release and inhibit cAMP production in the CB and that these effects are prevented by A2B agonists; conversely, D2 antagonists increase the release of catecholamines, this phenomenon being also prevented by A2B antagonists. A2B-D2 RRI, however, have not yet been demonstrated to date in any cell type.

Homo- and Hetero-Dimers Between Opioid Receptors (μOR and δOR)

μOR forms homodimers (Lopez and Salome, 2009) with an elevated stability, as demonstrated in transfected HEK-293 cells by BRET and radioligand binding assays (Wang et al., 2005). μOR/μOR homodimerization occurs prior of transportation to the cell membrane. Sarkar et al. (2012) also proved μOR homodimerization in rat NK cells through Western-blot analysis and immunoprecipitation as well as immunohistochemistry and immunofluorescence.

Also δOR homodimerization was confirmed by Western-blotting and co-immunoprecipitation in different transfected cellular systems, such as CHO cells and COS cells (Cvejic and Devi, 1997). Data obtained by cross-linking experiments in CHO cells indicated that homodimerization does not depend on the expression level of the receptor. Subsequently, the occurrence of homodimers was also assessed in HEK-293T cells by BRET (Johnston et al., 2011) and in rat NK cells by Western-blot assays, immunoprecipitation, immunohistochemistry, and immunofluorescence (Sarkar et al., 2012).

δOR/μOR heterodimers have also been demonstrated (George et al., 2000; Gomes et al., 2002; Sarkar et al., 2012). The heteroreceptor complexes exhibit distinct pharmacological properties from that of the monomers. Low non-signaling doses of δ- or μ-ligands potentiate the binding and signaling of the receptors and the affinity of endomorphin-1 and δ-selective agonists is increased in the heteroreceptor complex, when compared to monomers (Kabli et al., 2010). These properties appeared linked to changes in the signaling pathways activated by the heteroreceptor complexes as compared to individual receptors (Gomes et al., 2013). In NK cells, in which δOR and μOR proteins exist as homo- and heterodimers, differences in cell function were observed depending on the prevalent type of receptor dimerization (Sarkar et al., 2012).

As suggested by Cvejic and Devi (1997) the dimerization also plays an important role in the internalization of the δOR receptor which in its monomeric form may not be internalized. This could be related to the mechanism that takes place after long-term exposure to opioid ligands. Dimerization of δOR receptors seems to be agonist dependent.

As it regards the CB, both μOR and δOR are expressed in type I cells (Ichikawa et al., 2005; Ricker et al., 2015). The expression of both receptors in type I cells suggests the occurrence of homo- and hetero-dimerization, with possible pharmacological implications. Perivascular nerve fibers have also been reported to express δOR (Ichikawa et al., 2005) although further studies will be necessary for detection of other opioid receptor types.

Hetero-Dimers Between Dopamine (D1) and Opioid (μOR) Receptors

Co-immunoprecipitation, BRET and cross-antagonism assays demonstrated the existence of D1/μOR heterodimers in both transfected HEK-293 cells and ventral striatum (Tao et al., 2017). D1 receptor antagonist can antagonize μOR-mediated signaling and function in a dopamine-independent manner, mostly likely via allosteric interactions through the D1/μOR heteromer. D1 antagonist (SCH23390) prevented opiate-induced activation of G-protein, inhibition of adenylyl cyclase, phosphorylation of ERK 1/2 and expression of c-fos in transfected cells expressing both receptors and in striatal tissues from wild type, but not DR KO, mice. The ability of an antagonist to inhibit signals originated by stimulation of the partner receptor is a biochemical characteristic that has been described for receptor heterodimers (cross-antagonism) (Tao et al., 2017).

As discussed above, D1 receptor and μOR were both assessed in the CB, supporting the hypothesis of corresponding RRI. Moreover, changes in the dynamic monomer/oligomer equilibrium may be postulated on the basis of environmental stimuli, as D1 expression levels, for instance, decrease in hypoxia conditions and along with exposure time (Bairam et al., 2003).

Hetero-Dimers Between Dopamine (D1 and D2) and Histamine (H3) Receptors

Koerner et al. (2004) assessed the presence of histamine receptors (H1, H2, H3) in the CB through RT-PCR studies. In particular, H3 has been detected in rat type I cells by immunohistochemistry (Lazarov et al., 2009; Del Rio et al., 2009; Thompson et al., 2010). H3 antagonism results in increased chemosensory activity (Del Rio et al., 2009) and H3 activation inhibits the intracellular Ca2+ signaling mediated by activation of muscarinic receptors in type I cells (Thompson et al., 2010). On the basis of the above and following findings, histamine receptors could also be included in the series of GPCRs with potential oligomerization in the CB.

In fact, Ferrada et al. (2009) demonstrated the D1–H3 receptor heteromerization in mammalian transfected cells (HEK-293) by BRET and binding assays as well as in the neuronal cell model SK-N-MC by radioligand experiments. The presence of this heteromer in the brain (striatum) was then also proved by Moreno et al. (2011).

An antagonist acting on one of the receptor units that constitute the D1–H3 receptor heteromer can cause changes in conformation of the other receptor unit, thus, blocking specific signals that originate in the heteromer. This mechanism could be responsible for unsuspected GPCR antagonists-related therapeutic potentials (Ferrada et al., 2009). D1/H3 receptor heteromers work as processors integrating dopamine- as well as histamine-related signals involved in controlling the function of striatal neurons of the direct striatal pathway (Moreno et al., 2011).

A strong and selective heteromeric interaction between D2 and H3 was also assessed by radioligand binding experiments in striatal membrane preparations and in co-transfected HEK-293 cells by BRET (Ferrada et al., 2008). According to agonist/antagonist competition experiments, a significant decrease of D2 receptors affinity for the agonist was encountered after stimulation of H3 receptors with several H3 agonists (Ferrada et al., 2008).

Hetero-Dimers Between Dopamine (D2) and Serotonin (5-HT2A) Receptors

5-HT2A/D2 heteroreceptor complexes were demonstrated by PLA and immunofluorescence in co-transfected HEK-293T cells (Lukasiewicz et al., 2010; Borroto-Escuela et al., 2014), as well as in discrete regions of the ventral and dorsal striatum and nucleus accumbens (Borroto-Escuela et al., 2014), medial prefrontal cortex as well as in the pars reticulate of the substantia nigra in rat (Lukasiewicz et al., 2010).

The functional properties of this complex are not yet completely clear (Lukasiewicz et al., 2010). However, ligands with high 5-HT2A/D2 selectivity and partial agonistic activity on D2 have been recently identified as new antipsychotic drugs which do not significantly induce extrapyramidal side effects (Möller et al., 2015).

Serotonin is released by type I cells and it acts in a autocrine/paracrine manner on 5-HT2 receptors which are localized in type I cells, as proved by immunofluorescence and pharmacological/functional studies in the rat CB (Zhang et al., 2003; Jacono et al., 2005; Liu et al., 2011; Yokoyama et al., 2015). Possible expression of 5-HT2A in carotid sinus nerve terminals has also been reported (Nurse et al., 2018). Serotonin is also considered to be involved in long-term facilitation of CB sensory discharge due to chronic intermittent hypoxia (Peng et al., 2009; Prabhakar, 2011). The presence of 5-HT2A and D2 in type I cells suggests the possibility of heterodimer formation, with functional roles to be investigated. 5-HT2A and D2 mediate excitatory and inhibitory effects on type I cells and the role of dimerization in the modulation of the two response types is particularly intriguing.

Hetero-Dimers Between Dopamine (D2) and Neurotensin (NTS1) Receptors

Among CB receptors possibly interacting with D2 receptor there is also the neurotensin receptor 1 (NTS1), which has been demonstrated by immunohistochemistry in rat and human type I cells (Porzionato et al., 2009). D2-NTS1 complexes have also been highlighted in living HEK293T cells by BRET technology (Borroto-Escuela et al., 2013b). Through these RRI neurotensin reduces dopamine binding and function at D2 receptor, moving dopamine transmission toward D1 receptor, which is not antagonized by NTS1 (Borroto-Escuela and Fuxe, 2017).

Homo- and Hetero-Dimers Between Melatonin Receptors (MT1 and MT2)

MT2 receptors have the ability to form homodimers, as first demonstrated in HEK-293T transfected cells through BRET approaches (including single-cell BRET experiments), as well as Western-blotting and co-immunoprecipitation assays (Ayoub et al., 2002). However, it has also been observed that MT2 receptors tend to preferentially form heterodimers with MT1, as proved by BRET techniques (BRET donor saturation assay) in the same cell model (Ayoub et al., 2004).

The homodimerization of MT2 receptors is not modulated by ligands as it can exist in a constitutive manner in living cells. However, the speculated role of MT2 dimers is that they are required for explicating biological functions of cells being considered functional signaling units. Modulation of the signaling and trafficking occurs similarly to the GABAB receptors (Ayoub et al., 2002).

As regards the presence of MT receptors in the CB, an in situ hybridization study showed MT2 expression in the rat type I cells whereas there are not yet data about MT1 (Tjong et al., 2004). Thus, to date, we can suppose MT2 homodimerization in type I cells although further analyses could be of interest for MT1 detection.

Galanin Receptor (Gal1, Gal2) Heteromers

Galanin (Mazzatenta et al., 2014) and galanin receptors 1 (Gal1) and 2 (Gal2), but not 3 (Gal3), (Porzionato et al., 2010) have been demonstrated by immunohistochemistry in rat type I cells. Galanin is known to regulate the differentiation of neural stem cells and plasticity responses in the nervous system (e.g., Cordeo-Llana et al., 2014) and it has been suggested that galanin expression in chemoreceptor cells could provide a signal for neurogenesis and chemoreceptor cell differentiation (Di Giulio et al., 2015; Mazzatenta et al., 2016).

As it concerns this paper, galanin receptors are particularly intriguing because evidence is given about the possibility of Gal1-Gal2, D1-Gal1, Gal-NPYY1, Gal1-Gal2-NPYY2 or Gal1-Gal2-AT1 heteromers, which can be postulated in type I cells (reviewed in Fuxe et al., 2012b).

Although there are not specific data about the expression of the different NPY receptor types in the CB, NPY immunoreactivity has been identified in nerve fibers as well as type I cells of dog, monkey and rat CB (Oomori et al., 1991, 2002). In the rat CB, a reduction in NPY-immunoreactive type I cells was observed from postnatal week 2 onward; conversely, NPY-immunoreactive fibers mainly increase since week 2 after birth (Oomori et al., 2002).

The local renin-angiotensin system in the CB has been extensively studied in the past years (e.g., Fung et al., 2002; Fung, 2014; Lam et al., 2014) and angiotensin II receptor type 1 (AT1) has been identified in rat type I (Fung et al., 2001; Atanasova et al., 2018) and type II (Murali et al., 2014) cells. In the rat, AT1 receptors are also up-regulated in response to hypoxia (Fung et al., 2002).

Homo- and Hetero-Dimers Between Endothelin Receptors (ETA and ETB)

Endothelin 1 (ET-1) receptors type A (ETA) and B (ETB) may form homo- and heterodimers, although the functional implications of different dimerization are still largely unknown. Their behavior was studied after expression in transfected HEK-293T cells by immunoprecipitation, immunoblotting and FRET (Evans and Walker, 2008). These results confirmed the previous data by Gregan et al. (2004) who adopted FRET too, co-localizing ET-1 receptors at the plasma membrane. Furthermore, the latter speculated that ET-1-receptors homodimerization (as well as homo-oligomerization) is constitutive and ligand-independent (Evans and Walker, 2008).

Type I cells synthetize and release ET-1, which may act in autocrine/paracrine manner on the same type I cells through both the above receptors. Chronic continuous hypoxia upregulates ET-1 and ETA receptor, suggesting a critical role in chronic hypoxia-induced increased chemosensitivity in the rat CB (Chen et al., 2002). Chronic intermittent hypoxia is also known to enhance the CB chemosensory and ventilatory responses to acute hypoxia and produce long-term sensory potentiation of chemosensory discharges (e.g., Peng et al., 2003, 2013; Rey et al., 2004; Pawar et al., 2009). Chronic intermittent hypoxia has been reported to determine an upregulation of ET-1 and ETB receptor, but not of ETA receptor, in adult cats (Rey et al., 2007); conversely, upregulation of ET-1 and ETA receptor, but not ETB receptor, have been reported in neonatal (Pawar et al., 2009) and adult (Peng et al., 2013) rats.

Endothelin rise intracellular Ca2+ in cultured type II cells (Murali et al., 2015). Stimulation of ETB receptors by ET-1 has been reported to play a role in proliferation of stem cells derived from type II cells and CB hyperplasia following chronic hypoxia (Platero-Luengo et al., 2014).

Thus, RRIs between ET-1 receptors can be hypothesized in type I and II cells. Hypoxia-induced changes in the expression of two different receptor types, together with the involvement of ET-1 in functional/structural modifications, suggests the idea that changes in the monomer/(homo/hetero)-dimer equilibrium may also play a role.

ETB receptor has also been reported to undergo physical interactions with AT1 receptor located in the cells of renal proximal tubule (Zeng et al., 2005). RRI between the two receptors would be particularly intriguing in the CB, due to their roles in hypoxia responses.

Hetero-Dimers Between GABA (GABAB2) and Muscarinic (M2) Receptors

Boyer et al. (2009) demonstrated by FRET that GABAB2 is co-localized and directly associates with M2 receptor in neuronal PC12 cells. In parallel, the authors observed that in another cell model, i.e., HEK-293T, GABAB1 is also required for GABAB2/M2 heterodimerization, contrary to what was observed in PC12. Moreover, through co-immunoprecipitation and immunostaining experiments, the authors supported that signaling complexes GABAB2/M2 exist also in vivo in the brain cortex. The association seems to be specific since GABAB2 did not associate closely with other related muscarinic receptors (M1) or with a different GPCR (μOR).

The findings that M2 and GABAB2 are co-localized and associate in cortical neurons, which overlap with brain regions that receive cholinergic projections, suggest that the heterodimer is involved in a novel mechanism for enhancing cholinergic signaling in the brain. In fact, expression of GABAB2 in M2-expressing neurons would allow some neurons to maintain muscarinic signaling during elevated or chronic agonist exposure (Boyer et al., 2009).

Type I cells of the CB express both GABAB1 and GABAB2 subunits, as confirmed by RT-PCR studies. In addition to molecular biology evidences, localization of GABAB2 receptor subunits in sections of the CB was also assayed by immunofluorescence which assessed positive immunoreactivity for the receptor subunit in type I clusters (Fearon et al., 2003). Shirahata et al. (2004) demonstrated the expression of M2 mRNA and the presence of the receptor protein, by RT-PCR and immunohistochemistry, respectively. In particular, regarding localization, immunohistochemical analysis highlighted M2 presence in type I cells and petrosal afferent terminals. GABA and acetylcholine (Dasso et al., 1997) show inhibitory and excitatory actions, respectively, on type I cells. The possible occurrence of dimerization may have a role in the reciprocal modulation of actions of the two neurotransmitters.

Muscarinic receptors are also present in type II cells, as muscarinic agonists elicit an increase of intracellular Ca2+ levels in these cells (Tse et al., 2012; Murali et al., 2015), but there are not data about the possible expression of GABA receptors too.

Cannabinoid Receptor (CB1) Heteromers

The CB1 receptor was found to be expressed in the CB by techniques such as RT-PCR, immunohistochemistry and in situ hybridization, although its levels were relatively low (McLemore, 2004). These findings suggest that endocannabinoids may have an impact on blood flow regulation in the CB, therefore affecting the oxygen pressure and respiratory control. However, the CB1 function in CB needs to be further investigated, as data present in another study on CB1 were somewhat discording (Roy et al., 2012).

The CB1 can form heterodimers with many other different GPCRs which are also assessed to be present in the CB; among these δOR, μOR, A2A and D2 receptors. In particular, the ability of CB1 and D2 receptors to form heterodimers was demonstrated in co-transfected cells (HEK-293T) through co-immunoprecipitation and FRET techniques (Kearn, 2005; Marcellino et al., 2008). It is noteworthy that the activation of CB1-D2 receptor heterodimer can have completely opposite effects than activation of the individual receptors.

The above receptors have also been demonstrated to oligomerize in receptor mosaics so that the presence of these complexes may also be postulated for the CB. In particular, the existence of a CB1-D2-A2A receptor mosaic has been demonstrated, where CB1 receptor activation removes the D2 inhibition of the A2A receptor signaling (Fuxe et al., 2008; Marcellino et al., 2008). The above receptor mosaicism could represent a further way of subtle reciprocal modulation between different neurotransmitters.

Heterocomplexes Between GPCR and Other Receptor Types

G protein-coupled receptors may undergo direct interactions with other membrane receptors, such as ion channel receptors or receptor tyrosine kinases (Borroto-Escuela et al., 2016). Some of these complexes can be postulated in the CB, with possible roles in modulation of neurotransmission and plasticity.

For instance, NMDA receptor subunits 1, 2A and 2B have been detected in CB, through RT-PCR, and type I cells, through immunohistochemistry. Chronic intermittent hypoxia also increases the expression of NMDA1 and NMDA2B receptors (Liu et al., 2009). It is noteworthy that D1 receptors can regulate the function of the NMDA receptor by means of direct protein–protein interactions between the carboxyl terminals of D1 receptor and NMDA1 and NMDA2A (Lee et al., 2002; Li et al., 2010), stimulating NMDA receptor-mediated long-term potentiation (Nai et al., 2010).

FGF receptor 1, which is a tyrosine kinases receptor, has been reported to undergo direct RRI with A2A receptor (Flajolet et al., 2008; Borroto-Escuela et al., 2013a). In particular, as demonstrated in PC12 cells (showing a series of similar properties with type I cells), contemporary activation of the two receptors, but not the single ones, induces activation of the MAPK/ERK pathway, differentiation and neurite extension (Flajolet et al., 2008). The majority of human CB’s type I cells have shown a weak to moderate cytoplasmic immunostaining for FGF1 receptor (Douwes Dekker et al., 2007). In particular, FGF1 receptor immunoreactivity was shown in type I cells in postnatal rat CB cultures, in both normoxic and hypoxic conditions, as well as in bFGF presence or absence (Paciga and Nurse, 2001). bFGF increased both inward Na+ and outward K+ currents after 2 days of treatment on cultured type I cells from E18-19 rat pups (Zhong and Nurse, 1995). In cultures from rat E17-E19 CB, bFGF increases survival and BrdU incorporation; in postnatal P1-P3 cultures, bFGF still stimulates DNA synthesis but does not affect survival. In fetal rat glomus cells, bFGF stimulates neuronal differentiation, producing neurite outgrowth and inducing neurofilament immunoreactivity; these changes can no longer be detected in postnatal cultures (Nurse and Vollmer, 1997). Given the concomitant expression of A2A receptors in type I cells, an heterocomplex could be hypothesized. Moreover, the above developmental changes in the FGF action could partially derive by changes in the monomer–heteromer equilibrium. It is known, for instance, that the expression of A2A receptors in the rat type I cells decreases by PN14 (Gauda et al., 2000).

It is noteworthy that A2A-D2-FGF receptor mosaic has also been highlighted in other cell types (Fuxe et al., 2010) and could represent a further way of integration between neuromodulation and plasticity mechanisms.

Conclusion and Future Perspectives

The CB is characterized by the production and release of many different neurotransmitters and neuromodulators which act on various receptor types identifiable on type I and II cells and nerve fibers. Although the main receptors involved in conveying excitatory stimuli from type I cells to afferent nerve fibers are ionotropic (nicotinic, P2X2/P2X3), a wide series of GPCRs is also expressed in the various structures involved in chemoreception, exerting modulation of neurotransmission. In other transfected and native cell types, experimental evidence has been provided about the existence of RRIs between GPCRs, with production of homodimers, heterodimers or high-order complexes (receptor mosaics) which may modify ligand binding, signaling and internalization of the protomers. In the present paper, we have reviewed most literature data reporting the contemporary expression in type I cells, type II cells or afferents of GPCRs which are able to physically interact with each other to form homo/hetero-complexes. This is a prerequisite for postulating dimerization and/or oligomerization in the CB. RRIs are particularly intriguing to be hypothesized in the CB, where in vitro/in vivo pharmacological/functional data are available about cross-talks between different neurotransmitters/neuromodulators or ‘paradoxical’ response changes with different concentrations/doses. At least in some cases, such findings could be interpreted through the existence of direct RRIs. Nevertheless, few authors have considered the possibility of di/oligo-merization in the CB (Conde et al., 2008, 2009b) and to the best of our knowledge there are not studies addressing this aspect through up-to-date methodology (see corresponding paragraph). The literature data reviewed in the present paper support the possibility of RRI in the CB and stress the potential implications of di/oligomerization for chemoreception, neuromodulation and plasticity. It is known, in fact, that the expression of certain receptors varies in response to development or environmental stimuli (hypoxia, hyperoxia, etc.); changes in the dynamic monomer/oligomer equilibrium may be the consequence, with correlated functional implications.

In the present paper, we have collected indirect evidence of our ‘working hypothesis,’ identifying the most significant homo/hetero-complexes which would be worthwhile to be studied in the CB. The direct identification of homo/hetero-complexes in type I, type II and afferent terminations and the characterization of their functional role and relevance in the chemoreception could represent an exciting field of investigation. As previously stated, demonstration of RRIs in the CB should include (1) assessment of colocalization in “close proximity” of two or more receptors and (2) detectable biochemical/functional change in one receptor induced by the binding of a ligand to another receptor. We have here revised literature data highlighting colocalization of different receptors in the various elements of the CB (i.e., type I and II cells, nerve terminals). In addition, colocalization of other receptors could be preliminarily investigated through double immunofluorescence or in situ hybridization, both in CB tissue samples and cell cultures.

However, colocalization is just a necessary condition to have direct RRIs and the demonstration of close proximity (i.e., a distance lower than 10 nm) between receptor molecules must be provided. To date, many methods of both biochemical and biophysical nature are available to accomplish this task and most of them could be applied to CB cell cultures. In particular, BRET, FRET or atomic force microscopy could be considered the most useful approaches. Interestingly, in vitro models allow to detect changes in cultured CB cells as a response to external pharmacological or environmental (hypoxia, hyperoxia, etc.) stimuli; we have hypothesized that RRIs may increase or decrease in response to external actions. Dynamic modifications of RRIs could also be analyzed in living cells through experimental techniques such as total internal reflection fluorescence microscopy, fluorescence correlation spectroscopy and PALM. Apart from cell cultures, heterocomplexes could be identified in native formalin-fixed CBs through in situ PLA, an approach that would even permit the identification of eventual changes in RRIs with respect to experimental conditions of in vivo models.

Once demonstrated physical RRIs, functional approaches in cell cultures would have to verify how the agonist/antagonist to one receptor may modify the response of the other receptor(s) toward the corresponding agonists/antagonists.

In conclusion, many in vitro and in vivo models are available, for research in CB structure/function, which represent adequate material for the application of consistent methods of analysis of potential RRIs.

Author Contributions

All the authors contributed to the revision of the literature and discussion of the hypothesis proposed. All the authors read and approved the final version of the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Agnati, L. F., Ferré, S., Lluis, C., Franco, R., and Fuxe, K. (2003). Molecular mechanisms and therapeutical implications of intramembrane receptor/receptor interactions among heptahelical receptors with examples from the striatopallidal GABA neurons. Pharmacol. Rev. 55, 509–550. doi: 10.1124/pr.55.3.2

PubMed Abstract | CrossRef Full Text | Google Scholar

Agnati, L. F., Fuxe, K., Torvinen, M., Genedani, S., Franco, R., Watson, S., et al. (2005). New methods to evaluate colocalization of fluorophores in immunocytochemical preparations as exemplified by a study on A2A and D2 receptors in Chinese hamster ovary cells. J. Histochem. Cytochem. 53, 941–953. doi: 10.1369/jhc.4A6355.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

Agnati, L. F., Guidolin, D., Leo, G., Carone, C., Genedani, S., and Fuxe, K. (2010). Receptor-receptor interactions: a novel concept in brain integration. Prog. Neurobiol. 90, 157–175. doi: 10.1016/j.pneurobio.2009.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Almaraz, L., Perez-Garcia, M. T., and Gonzalez, C. (1991). Presence of D1 receptors in the rabbit carotid body. Neurosci. Lett. 132, 259–262. doi: 10.1016/0304-3940(91)90315-K

CrossRef Full Text | Google Scholar

Atack, J. R., Shook, B. C., Rassnick, S., Jackson, P. F., Rhodes, K., Drinkenburg, W. H., et al. (2014). JNJ-40255293, a novel adenosine A2A/A1 antagonist with efficacy in preclinical models of Parkinson’s disease. ACS Chem. Neurosci. 5, 1005–1019. doi: 10.1021/cn5001606

PubMed Abstract | CrossRef Full Text | Google Scholar

Atanasova, D. Y., Dandov, A. D., Dimitrov, N. D., and Lazarov, N. E. (2018). Immunohistochemical localization of angiotensin AT1 receptors in the rat carotid body. Acta Histochem. 120, 154–158. doi: 10.1016/j.acthis.2018.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Atanasova, D. Y., and Lazarov, N. E. (2014). Expression of neurotrophic factors and their receptors in the carotid body of spontaneously hypertensive rats. Respir. Physiol. Neurobiol. 202, 6–15. doi: 10.1016/j.resp.2014.06.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Ayoub, M. A., Couturier, C., Lucas-Meunier, E., Angers, S., Fossier, P., Bouvier, M., et al. (2002). Monitoring of ligand-independent dimerization and ligand-induced conformational changes of melatonin receptors in living cells by bioluminescence resonance energy transfer. J. Biol. Chem. 277, 21522–21528. doi: 10.1074/jbc.M200729200

PubMed Abstract | CrossRef Full Text | Google Scholar

Ayoub, M. A., Delagrange, P., and Jockers, R. (2004). Preferential formation of MT 1 / MT 2 melatonin receptor heterodimers with distinct ligand interaction properties. Mol. Pharmacol. 66, 312–321. doi: 10.1124/mol.104.000398.reasonable

PubMed Abstract | CrossRef Full Text | Google Scholar

Bae, H., Nantwi, K. D., and Goshgarian, H. G. (2005). Recovery of respiratory function following C2 hemi and carotid body denervation in adult rats: influence of peripheral adenosine receptors. Exp. Neurol. 191, 94–103. doi: 10.1016/j.expneurol.2004.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Bairam, A., and Carroll, J. L. (2005). Neurotransmitters in carotid body development. Respir. Physiol. Neurobiol. 149, 217–232. doi: 10.1016/j.resp.2005.04.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Bairam, A., Carroll, J. L., Labelle, Y., and Khandjian, E. W. (2003). Differential changes in dopamine D2- and D1-receptor mRNA levels induced by hypoxia in the arterial chemoreflex pathway organs in one-day-old and adult rabbits. Biol. Neonate 84, 222–231. doi: 10.1159/000072306

PubMed Abstract | CrossRef Full Text | Google Scholar

Bairam, A., Frenette, J., Dauphin, C., Carroll, J. L., and Khandjian, E. W. (1998). Expression of dopamine D1-receptor mRNA in the carotid body of adult rabbits, cats and rats. Neurosci. Res. 31, 147–154. doi: 10.1016/S0168-0102(98)00033-39

PubMed Abstract | CrossRef Full Text | Google Scholar

Bairam, A., Joseph, V., Lajeunesse, Y., and Kinkead, R. (2009). Altered expression of adenosine A1 and A2Areceptors in the carotid body and nucleus tractus solitarius of adult male and female rats following neonatal caffeine treatment. Brain Res. 1287, 74–83. doi: 10.1016/j.brainres.2009.06.064

PubMed Abstract | CrossRef Full Text | Google Scholar

Bayburt, T. H., Leitz, A. J., Xie, G., Oprian, D. D., and Sligar, S. G. (2007). Transducin activation by nanoscale lipid bilayers containing one and two rhodopsins. J. Biol. Chem. 282, 14875–14881. doi: 10.1074/jbc.M701433200

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonaventura, J., Navarro, G., Casadó-Anguera, V., Azdad, K., Rea, W., Moreno, E., et al. (2015). Allosteric interactions between agonists and antagonists within the adenosine A2A receptor-dopamine D2 receptor heterotetramer. Proc. Natl. Acad. Sci. 112, E3609–E3618. doi: 10.1073/pnas.1507704112

PubMed Abstract | CrossRef Full Text | Google Scholar

Borroto-Escuela, D. O., Flajolet, M., Agnati, L. F., Greengard, P., and Fuxe, K. (2013a). Bioluminescence resonance energy transfer methods to study G protein-coupled receptor-receptor tyrosine kinase heteroreceptor complexes. Methods Cell Biol. 117, 141–164. doi: 10.1016/B978-0-12-408143-7.00008-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Borroto-Escuela, D. O., Ravani, A., Tarakanov, A. O., Brito, I., Narvaez, M., Romero-Fernandez, W., et al. (2013b). Dopamine D2 receptor signaling dynamics of dopamine D2-neurotensin 1 receptor heteromers. Biochem. Biophys. Res. Commun. 435, 140–146. doi: 10.1016/j.bbrc.2013.04.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Borroto-Escuela, D. O., and Fuxe, K. (2017). Diversity and bias through dopamine D2R heteroreceptor complexes. Curr. Opin. Pharmacol. 32, 16–22. doi: 10.1016/j.coph.2016.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Borroto-Escuela, D. O., Romero-Fernandez, W., Narvaez, M., Oflijan, J., Agnati, L. F., and Fuxe, K. (2014). Hallucinogenic 5-HT2AR agonists LSD and DOI enhance dopamine D2R protomer recognition and signaling of D2-5-HT2A heteroreceptor complexes. Biochem. Biophys. Res. Commun. 443, 278–284. doi: 10.1016/j.bbrc.2013.11.104

PubMed Abstract | CrossRef Full Text | Google Scholar

Borroto-Escuela, D. O., Wydra, K., Pintsuk, J., Narvaez, M., Corrales, F., Zaniewska, M., et al. (2016). Understanding the functional plasticity in neural networks of the basal ganglia in cocaine use disorder: a role for allosteric receptor-receptor interactions in A2A-D2 heteroreceptor complexes. Neural Plast. 2016:4827268. doi: 10.1155/2016/4827268

PubMed Abstract | CrossRef Full Text | Google Scholar

Bouvier, M., and Hébert, T. E. (2014). CrossTalk proposal: Weighing the evidence for Class A GPCR dimers, the evidence favours dimers. J. Physiol. 592, 2439–2441. doi: 10.1113/jphysiol.2014.272252

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyer, S. B., Clancy, S. M., Terunuma, M., Revilla-Sanchez, R., Thomas, S. M., Moss, S. J., et al. (2009). Direct interaction of GABAB receptors with M2 muscarinic receptors enhances muscarinic signaling. J. Neurosci. 29, 15796–15809. doi: 10.1523/JNEUROSCI.4103-09.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Bulenger, S., Marullo, S., and Bouvier, M. (2005). Emerging role of homo- and heterodimerization in G-protein-coupled receptor biosynthesis and maturation. Trends Pharmacol. Sci. 26, 131–137. doi: 10.1016/j.tips.2005.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Calver, A. R., Medhurst, A. D., Robbins, M. J., Charles, K. J., Evans, M. L., Harrison, D. C., et al. (2000). The expression of GABA(B1) and GABA(B2) receptor subunits in the cNS differs from that in peripheral tissues. Neuroscience 100, 155–170. doi: 10.1016/S0306-4522(00)00262-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Canals, M., Burgueño, J., Marcellino, D., Cabello, N., Canela, E. I., Mallol, J., et al. (2004). Homodimerization of adenosine A2A receptors: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J. Neurochem. 88, 726–734. doi: 10.1046/j.1471-4159.2003.02200.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Canals, M., Marcellino, D., Fanelli, F., Ciruela, F., de Benedetti, P., Goldberg, S. R., et al. (2003). Adenosine A2A-dopamine D2 receptor-receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J. Biol. Chem. 278, 46741–46749. doi: 10.1074/jbc.M306451200

PubMed Abstract | CrossRef Full Text | Google Scholar

Carroll, J. L., Agarwal, A., Donnelly, D. F., and Kim, I. (2012). Purinergic modulation of carotid body glomus cell hypoxia response during postnatal maturation in rats. Adv. Exp. Med. Biol. 758, 249–253. doi: 10.1007/978-94-007-4584-1_34

PubMed Abstract | CrossRef Full Text | Google Scholar

Changeux, J. P., and Christopoulos, A. (2016). Allosteric modulation as a unifying mechanism for receptor function and modulation. Cell 166, 1084–1102. doi: 10.1016/j.cell.2016.08.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., He, L., Dinger, B., Stensaas, L., and Fidone, S. (2002). Role of endothelin and endothelin A-type receptor in adaptation of the carotid body to chronic hypoxia. Am. J. Physiol. Lung Cell. Mol. Physiol. 282, L1314–L1323. doi: 10.1152/ajplung.00454.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Wei, L. N., and Müller, J. D. (2003). Probing protein oligomerization in living cells with fluorescence fluctuation spectroscopy. Proc. Natl. Acad. Sci. U.S.A. 100, 15492–15497. doi: 10.1073/pnas.2533045100

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciruela, F., Casadó, V., Mallol, J., Canela, E. I., Lluis, C., and Franco, R. (1995). Immunological identification of A1 adenosine receptors in brain cortex. J. Neurosci. Res. 42, 818–828. doi: 10.1002/jnr.490420610

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciruela, F., Ferre, S., Casado, V., Cortes, A., Cunha, R. A., Lluis, C., et al. (2006). Heterodimeric adenosine receptors: a device to regulate neurotransmitter release. Cell. Mol. Life Sci. 63, 2427–2431. doi: 10.1007/s00018-006-6216-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Conde, S. V., Gonzalez, C., Batuca, J. R., Monteiro, E. C., and Obeso, A. (2008). An antagonistic interaction between A2B adenosine and D2 dopamine receptors modulates the function of rat carotid body chemoreceptor cells. J. Neurochem. 107, 1369–1381. doi: 10.1111/j.1471-4159.2008.05704.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Conde, S. V., Monteiro, E. C., Obeso, A., and Gonzalez, C. (2009a). Adenosine in peripheral chemoreception: new insights into a historically overlooked molecule. Adv. Exp. Med. Biol. 648, 145–159. doi: 10.1007/978-90-481-2259-2_17

PubMed Abstract | CrossRef Full Text | Google Scholar

Conde, S. V., Obeso, A., Monteiro, E. C., and Gonzalez, C. (2009b). The A(2B)-D(2) receptor interaction that controls carotid body catecholamines release locates between the last two steps of hypoxic transduction cascade. Adv. Exp. Med. Biol. 648, 161–168. doi: 10.1007/978-90-481-2259-2_18

PubMed Abstract | CrossRef Full Text | Google Scholar

Conde, S. V., Monteiro, E. C., Rigual, R., Obeso, A., and Gonzalez, C. (2012). Hypoxic intensity: a determinant for the contribution of ATP and adenosine to the genesis of carotid body chemosensory activity. J. Appl. Physiol. 112, 2002–2010. doi: 10.1152/japplphysiol.01617.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

Conde, S. V., Obeso, A., Vicario, I., Rigual, R., Rocher, A., and Gonzalez, C. (2006). Caffeine inhibition of rat carotid body chemoreceptors is mediated by A2A and A2B adenosine receptors. J. Neurochem. 98, 616–628. doi: 10.1111/j.1471-4159.2006.03912.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cordeo-Llana, O., Rinaldi, F., Brennan, P. A., Wynick, D., and Caldwell, M. A. (2014). Galanin promotes neuronal differentiation from neuronal progenitor cells in vitro and contributes to the generation of new olfactory neurons in the adult mouse brain. Exp. Neurol. 256, 93–104. doi: 10.1016/j.expneurol.2014.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Cvejic, S., and Devi, L. A. (1997). Dimerization of the delta opioid receptor: implication for a role in receptor internalization. J. Biol. Chem. 272, 26959–26964. doi: 10.1074/jbc.272.43.26959

CrossRef Full Text | Google Scholar

Dasso, L. L., Buckler, K. J., and Vaughan-Jones, R. D. (1997). Muscarinic and nicotinic receptors raise intracellular Ca2+ levels in rat carotid body type I cells. J. Physiol. 498, 327–338. doi: 10.1113/jphysiol.1997.sp021861

PubMed Abstract | CrossRef Full Text | Google Scholar

De Caro, R., Macchi, V., Sfriso, M. M., and Porzionato, A. (2013). Structural and neurochemical changes in the maturation of the carotid body. Respir. Physiol. Neurobiol. 185, 9–19. doi: 10.1016/j.resp.2012.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Del Rio, R., Moya, E. A., Alcayaga, J., and Iturriaga, R. (2009). Evidence for histamine as a new modulator of carotid body chemoreception. Adv. Exp. Med. Biol. 648, 177–184. doi: 10.1007/978-90-481-2259-2_20

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Giulio, C., Marconi, G. D., Zara, S., Di Tano, A., Porzionato, A., Pokorski, M., et al. (2015). Selective expression of galanin in neuronal-like cells of the human carotid body. Adv. Exp. Med. Biol. 860, 315–323. doi: 10.1007/978-3-319-18440-1_36

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Giulio, C., Zara, S., Cataldi, A., Porzionato, A., Pokorski, M., and De Caro, R. (2012). Human carotid body HIF and NGB expression during human development and aging. Adv. Exp. Med. Biol. 758, 265–271. doi: 10.1007/978-94-007-4584-1_36

PubMed Abstract | CrossRef Full Text | Google Scholar

Douwes Dekker, P. B., Kuipers-Dijkshoornb, N. J., Baelde, H. J., van der Mey, A. G. L., Hogendoorn, P. C. W., and Cornelisse, C. J. (2007). Basic fibroblast growth factor and fibroblastic growth factor receptor–1 may contribute to head and neck paraganglioma development by an autocrine or paracrine mechanism. Hum. Pathol. 38, 79–85. doi: 10.1016/j.humpath.2006.06.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Doyle, S. E., Breslin, F. J., Rieger, J. M., Beauglehole, A., and Lynch, W. J. (2012). Time and sex-dependent effects of an adenosine A2A/A1 receptor antagonist on motivation to self-administer cocaine in rats. Pharmacol. Biochem. Behav. 102, 257–263. doi: 10.1016/j.pbb.2012.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Evans, N. J., and Walker, J. W. (2008). Endothelin receptor dimers evaluated by FRET, ligand binding, and calcium mobilization. Biophys. J. 95, 483–492. doi: 10.1529/biophysj.107.119206

PubMed Abstract | CrossRef Full Text | Google Scholar

Farran, B. (2017). An update on the physiological and therapeutic relevance of GPCR oligomers. Pharmacol. Res. 117, 303–327. doi: 10.1016/j.phrs.2017.01.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Fearon, I. M., Zhang, M., Vollmer, C., and Nurse, C. A. (2003). GABA mediates autoreceptor feedback inhibition in the rat carotid body via presynaptic GABA B receptors and TASK-1. J. Physiol. 553, 83–94. doi: 10.1113/jphysiol.2003.048298

PubMed Abstract | CrossRef Full Text | Google Scholar

Fernández-Dueñas, V., Llorente, J., Gandía, J., Borroto-Escuela, D. O., Agnati, L. F., Tasca, C. I., et al. (2012). Fluorescence resonance energy transfer-based technologies in the study of protein-protein interactions at the cell surface. Methods 57, 467–472. doi: 10.1016/j.ymeth.2012.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Fernández-Dueñas, V., Pérez-Arévalo, A., Altafaj, X., Ferré, S., and Ciruela, F. (2017). Adenosine A1-A2A receptor heteromer as a possible target for early-onset Parkinson’s disease. Front. Neurosci. 11:652. doi: 10.3389/fnins.2017.00652

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferrada, C., Ferre, S., Casado, V., Cortes, A., Justinova, Z., Barnes, C., et al. (2008). Interactions between histamine H3 and dopamine D2 receptors and the implications for striatal function. Neuropharmacology 55, 190–197. doi: 10.1016/j.neuropharm.2008.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferrada, C., Moreno, E., Casadó, V., Bongers, G., Cortés, A., Mallol, J., et al. (2009). Marked changes in signal transduction upon heteromerization of dopamine D1and histamine H3receptors. Br. J. Pharmacol. 157, 64–75. doi: 10.1111/j.1476-5381.2009.00152.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferré, S., Casado, V., Devi, L. A., Filizola, M., Jockers, R., Lohse, M. J., et al. (2014). G protein-coupled receptor oligomerization revisited: functional and pharmacological perspectives. Pharmacol. Rev. 66, 413–434. doi: 10.1124/pr.113.008052

PubMed Abstract | CrossRef Full Text | Google Scholar

Ferré, S., Ciruela, F., Borycz, J., Solinas, M., Quarta, D., Antoniou, K., et al. (2008). Adenosine A1-A2A receptor heteromers: new targets for caffeine in the brain. Front. Biosci. 13, 2391–2399. doi: 10.2741/2852

PubMed Abstract | CrossRef Full Text | Google Scholar

Fink, J. S., Weaver, D. R., Rivkees, S. A., Peterfreund, R. A., Pollack, A. E., Adler, E. M., et al. (1992). Molecular cloning of the rat A2 adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Brain Res. Mol. Brain Res. 14, 186–195. doi: 10.1016/0169-328X(92)90173-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Flajolet, M., Wang, Z., Futter, M., Shen, W., Nuangchamnong, N., Bendor, J., et al. (2008). FGF acts as a co-transmitter through adenosine A(2A) receptor to regulate synaptic plasticity. Nat. Neurosci. 11, 1402–1409. doi: 10.1038/nn.2216

PubMed Abstract | CrossRef Full Text | Google Scholar

Foord, S. M. (2002). Receptor classification: post genome. Curr. Opin. Pharmacol. 2, 561–566. doi: 10.1016/S1471-4892(02)00214-X

CrossRef Full Text | Google Scholar

Franco, R., Martínez-Pinilla, E., Lanciego, J. L., and Navarro, G. (2016). Basic pharmacological and structural evidence for class A G-protein-coupled receptor heteromerization. Front. Pharmacol. 7:76. doi: 10.3389/fphar.2016.00076

PubMed Abstract | CrossRef Full Text | Google Scholar

Fung, M. L. (2014). The role of local renin-angiotensin system in arterial chemoreceptors in sleep-breathing disorders. Front. Physiol. 5:336. doi: 10.3389/fphys.2014.00336

PubMed Abstract | CrossRef Full Text | Google Scholar

Fung, M. L., Lam, S. Y., Chen, Y., Dong, X., and Leung, P. S. (2001). Functional expression of angiotensin II receptors in type-I cells of the rat carotid body. Pflugers. Arch. 441, 474–480. doi: 10.1007/s004240000445

CrossRef Full Text | Google Scholar

Fung, M. L., Lam, S. Y., Dong, X., Chen, Y., and Leung, P. S. (2002). Postnatal hypoxemia increases angiotensin II sensitivity and up-regulates AT1a angiotensin receptors in rat carotid body chemoreceptors. J. Endocrinol. 173, 305–313. doi: 10.1677/joe.0.1730305

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuxe, K., Borroto-Escuela, D. O., Marcellino, D., Romero-Fernandez, W., Frankowska, M., Guidolin, D., et al. (2012a). GPCR heteromers and their allosteric receptor-receptor interactions. Curr. Med. Chem. 19, 356–363. doi: 10.2174/092986712803414259

CrossRef Full Text | Google Scholar

Fuxe, K., Borroto-Escuela, D. O., Romero-Fernandez, W., Tarakanov, A. O., Calvo, F., Garriga, P., et al. (2012b). On the existence and function of galanin receptor heteromers in the central nervous system. Front. Endocrinol. 3:127. doi: 10.3389/fendo.2012.00127

CrossRef Full Text | Google Scholar

Fuxe, K., Ferré, S., Zoli, M., and Agnati, L. F. (1998). Integrated events in central dopamine transmission as analyzed at multiple levels. Evidence for intramembrane adenosine A2A/dopamine D2 and adenosine A1/dopamine D1 receptor interactions in the basal ganglia. Brain Res. Brain Res. Rev. 26, 258–273. doi: 10.1016/S0165-0173(97)00049-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuxe, K., Marcellino, D., Borroto-Escuela, D. O., Frankowska, M., Ferraro, L., Guidolin, D., et al. (2010). The changing world of G protein-coupled receptors: from monomers to dimers and receptor mosaics with allosteric receptor-receptor interactions. J. Recept. Signal Transduct. Res. 30, 272–283. doi: 10.3109/10799893.2010.506191

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuxe, K., Marcellino, D., Guidolin, D., Woods, A. S., and Agnati, L. F. (2008). Heterodimers and receptor mosaics of different types of G-protein-coupled receptors. Physiology 23, 322–332. doi: 10.1152/physiol.00028.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Gahbauer, S., and Böckmann, R. A. (2016). Membrane-mediated oligomerization of G protein coupled receptors and its implication for GPCR function. Front. Physiol. 7:494. doi: 10.3389/fphys.2016.00494

CrossRef Full Text | Google Scholar

Galvez, T., Duthey, B., Kniazeff, J., Blahos, J., Rovelli, G., Bettler, B., et al. (2001). Allosteric interactions between GB1 and GB2 subunits are required for optimal GABA(B) receptor function. EMBO J. 20, 2152–2159. doi: 10.1093/emboj/20.9.2152

PubMed Abstract | CrossRef Full Text | Google Scholar

Gandia, J., Galino, J., Amaral, O. B., Soriano, A., Lluís, C., Franco, R., et al. (2008). Detection of higher-order G protein-coupled receptor oligomers by a combined BRET-BiFC technique. FEBS Lett. 582, 2979–2984. doi: 10.1016/j.febslet.2008.07.045

PubMed Abstract | CrossRef Full Text | Google Scholar

Gauda, E. B. (2000). Expression and localization of A2a and A1-adenosine receptor genes in the rat carotid body and petrosal ganglia. A2a and A1-adenosine receptor mRNAs in the rat carotid body. Adv. Exp. Med. Biol. 475, 549–558.

PubMed Abstract | Google Scholar

Gauda, E. B., Cooper, R. Z., Donnelly, D. F., Mason, A., and McLemore, G. L. (2006). The effect of development on the pattern of A1 and A2a-adenosine receptor gene and protein expression in rat peripheral arterial chemoreceptors. Adv. Exp. Med. Biol. 580, 121–129. doi: 10.1007/0-387-31311-7_19

PubMed Abstract | CrossRef Full Text | Google Scholar

Gauda, E. B., Northington, F. J., Linden, J., and Rosin, D. L. (2000). Differential expression of a(2a), A(1)-adenosine and D(2)-dopamine receptor genes in rat peripheral arterial chemoreceptors during postnatal development. Brain Res. 872, 1–10. doi: 10.1016/S0006-8993(00)02314-2313

PubMed Abstract | CrossRef Full Text | Google Scholar

George, S. R., Fan, T., Xie, Z., Tse, R., Tam, V., Varghese, G., et al. (2000). Oligomerization of mu- and delta-opioid receptors. Generation of novel functional properties. J. Biol. Chem. 275, 26128–26135. doi: 10.1074/jbc.M000345200

PubMed Abstract | CrossRef Full Text | Google Scholar

George, S. R., O’Dowd, B. F., and Lee, S. P. (2002). G-protein-coupled receptor oligomerization and its potential for drug discovery. Nat. Rev. Drug Discov. 1, 808–820. doi: 10.1038/nrd913

PubMed Abstract | CrossRef Full Text | Google Scholar

Gines, S., Hillion, J., Torvinen, M., Le Crom, S., Casado, V., Canela, E. I., et al. (2000). Dopamine D1 and adenosine A1 receptors form functionally interacting heteromeric complexes. Proc. Natl. Acad. Sci. 97, 8606–8611. doi: 10.1073/pnas.150241097

PubMed Abstract | CrossRef Full Text | Google Scholar

Gomes, I., Ayoub, M. A., Fujita, W., Jaeger, W. C., Pfleger, K. D., and Devi, L. A. (2016). G protein-coupled receptor heteromers. Annu. Rev. Pharmacol. Toxicol. 56, 403–425. doi: 10.1146/annurev-pharmtox-011613-135952

PubMed Abstract | CrossRef Full Text | Google Scholar

Gomes, I., Filipovska, J., Jordan, B. A., and Devi, L. A. (2002). Oligomerization of opioid receptors. Methods 27, 358–365. doi: 10.1016/S1046-2023(02)00094-4

CrossRef Full Text | Google Scholar

Gomes, I., Fujita, W., Chandrakala, M. V., and Devi, L. A. (2013). Disease-specific heteromerization of G-protein-coupled receptors that target drugs of abuse. Prog. Mol. Biol. Transl. Sci. 117, 207–265. doi: 10.1016/B978-0-12-386931-9.00009-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Gracia, E., Cortés, A., Meana, J. J., García-Sevilla, J., Herhsfield, M. S., Canel, E. I., et al. (2008). Human adenosine deaminase as an allosteric modulator of human A1 adenosine receptor: abolishment of negative cooperativity for [3H](R)-pia binding to the caudate nucleus. J. Neurochem. 107, 161–170. doi: 10.1111/j.1471-4159.2008.05602.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Gracia, E., Moreno, E., Cortés, A., Lluís, C., Mallol, J., McCormick, P. J., et al. (2013). Homodimerization of adenosine A1 receptors in brain cortex explains the biphasic effects of caffeine. Neuropharmacology 71, 56–69. doi: 10.1016/j.neuropharm.2013.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Gregan, B., Schaefer, M., Rosenthal, W., and Oksche, A. (2004). Fluorescence resonance energy transfer analysis reveals the existence of endothelin-A and endothelin-B receptor homodimers. J. Cardiovasc. Pharmacol. 44, 30–33. doi: 10.1097/01.fjc.0000166218.35168.79

PubMed Abstract | CrossRef Full Text | Google Scholar

Grisshammer, R. (2017). New approaches towards the understanding of integral membrane proteins: a structural perspective on G protein-coupled receptors. Protein Sci. 26, 1493–1504. doi: 10.1002/pro.3200

PubMed Abstract | CrossRef Full Text | Google Scholar

Guescini, M., Leo, G., Genedani, S., Carone, C., Pederzoli, F., Ciruela, F., et al. (2012). Microvesicle and tunneling nanotube mediated intracellular transfer of G-protein coupled receptors in cell cultures. Exp. Cell Res. 318, 603–613. doi: 10.1016/j.yexcr.2012.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Guidolin, D., Agnati, L. F., Marcoli, M., Borroto-Escuela, D., and Fuxe, K. (2015). G-protein-coupled receptor type A heteromers as an emerging therapeutic target. Expert Opin. Ther. Targets 19, 265–283. doi: 10.1517/14728222.2014.981155

PubMed Abstract | CrossRef Full Text | Google Scholar

Guidolin, D., Marcoli, M., Tortorella, C., Maura, G., and Agnati, L. F. (2018). G protein-coupled receptor-receptor interactions give integrative dynamics to intercellular communication. Rev. Neurosci. doi: 10.1515/revneuro-2017-0087 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Gurevich, V. V., and Gurevich, E. V. (2008). How and why do GPCRs dimerize? Trends Pharmacol. Sci. 29, 234–240. doi: 10.1016/j.tips.2008.02.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasbi, A., Perreault, M. L., Shen, M. Y. F., Fan, T., Nguyen, T., Alijaniaram, M., et al. (2018). Activation of dopamine D1-D2 receptor complex attenuates cocaine reward and reinstatement of cocaine-seeking through inhibition of DARPP-32, ERK, and ΔFosB. Front. Pharmacol. 8:924. doi: 10.3389/fphar.2017.00924

PubMed Abstract | CrossRef Full Text | Google Scholar

Hern, J. A., Baig, A. H., Mashanov, G. I., Birdsall, B., Corrie, J. E., Lazareno, S., et al. (2010). Formation and dissociation of M1 muscarinic receptor dimers seen by total internal reflection fluorescence imaging of single molecules. Proc. Natl. Acad. Sci. U.S.A. 107, 2693–2698. doi: 10.1073/pnas.0907915107

PubMed Abstract | CrossRef Full Text | Google Scholar

Hillion, J., Canals, M., Torvinen, M., Casadó, V., Scott, R., Terasmaa, A., et al. (2002). Coaggregation, cointernalization, and codesensitization of adenosine A2A receptors and dopamine D2 receptors. J. Biol. Chem. 277, 18091–18097. doi: 10.1074/jbc.M107731200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hopf, F. W., Cascini, M. G., Gordon, A. S., Diamond, I., and Bonci, A. (2003). Cooperative activation of dopamine D1 and D2 receptors increases spike firing of nucleus accumbens neurons via G-protein betagamma subunits. J. Neurosci. 23, 5079–5087. doi: 10.1523/JNEUROSCI.23-12-05079.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Huey, K. A., and Powell, F. L. (2000). Time-dependent changes in dopamine D2-receptor mRNA in the arterial chemoreflex pathway with chronic hypoxia. Mol. Brain Res. 75, 264–270. doi: 10.1016/S0169-328X(99)00321-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ichikawa, H., Schulz, S., Höllt, V., and Sugimoto, T. (2005). Delta-opioid receptor-immunoreactive neurons in the rat cranial sensory ganglia. Brain Res. 1043, 225–230. doi: 10.1016/j.brainres.2005.02.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Iturriaga, R., and Alcayaga, J. (2004). Neurotransmission in the carotid body: transmitters and modulators between glomus cells and petrosal ganglion nerve terminals. Brain Res. Brain Res. Rev. 47, 46–53. doi: 10.1016/j.brainresrev.2004.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacono, F. J., Peng, Y. J., Kumar, G. K., and Prabhakar, N. R. (2005). Modulation of the hypoxic sensory response of the carotid body by 5-hydroxytryptamine: role of the 5-HT2 receptor. Respir. Physiol. Neurobiol. 145, 135–142. doi: 10.1016/j.resp.2004.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnston, J. M., Aburi, M., Provasi, D., Bortolato, A., Urizar, E., Lambert, N. A., et al. (2011). Making structural sense of dimerization interfaces of delta opioid receptor homodimers. Biochemistry 50, 1682–1690. doi: 10.1021/bi101474v

PubMed Abstract | CrossRef Full Text | Google Scholar

Jonas, K. C., Huhtaniemi, I., and Hanyaloglu, A. C. (2016). Single-molecule resolution of G protein-coupled receptor (GPCR) complexes. Methods Cell Biol. 132, 55–72. doi: 10.1016/bs.mcb.2015.11.005-

PubMed Abstract | CrossRef Full Text | Google Scholar

Kabli, N., Martin, N., Fan, T., Nguyen, T., Hasbi, A., Balboni, G., et al. (2010). Agonists at the δ-opioid receptor modify the binding of μ-receptor agonists to the μ-δ receptor hetero-oligomer. Br. J. Pharmacol. 161, 1122–1136. doi: 10.1111/j.1476-5381.2010.00944.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaczor, A. A., and Selent, J. (2011). Oligomerization of G protein-coupled receptors: biochemical and biophysical methods. Curr. Med. Chem. 18, 4606–4634. doi: 10.2174/092986711797379285

CrossRef Full Text | Google Scholar

Kamal, M., and Jockers, R. (2011). Biological significance of GPCR heteromerization in the neuro-endocrine system. Front. Endocrinol. 2:2. doi: 10.3389/fendo.2011.00002

CrossRef Full Text | Google Scholar

Kearn, C. S. (2005). Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: a mechanism for receptor cross-talk? Mol. Pharmacol. 67, 1697–1704. doi: 10.1124/mol.104.006882

PubMed Abstract | CrossRef Full Text | Google Scholar

Kenakin, T., Agnati, L. F., Caron, M., Fredholm, B., Guidolin, D., Kobilka, B., et al. (2010). International workshop at the Nobel Forum, Karolinska Institutet on G protein-coupled receptors: finding the words to describe monomers, oligomers, and their molecular mechanisms and defining their meaning. Can a consensus be reached? J. Recep. Signal Transduct. Res. 30, 284–286. doi: 10.3109/10799893.2010.512438

PubMed Abstract | CrossRef Full Text | Google Scholar

Kleinau, G., Muller, A., and Biebermann, H. (2016). Oligomerization of GPCRs involved in endocrine regulation. J. Mol. Endocrinol. 57, R59–R80. doi: 10.1530/JME-16-0049

PubMed Abstract | CrossRef Full Text | Google Scholar

Kniazeff, J., Prezeau, L., Rondard, P., Pin, J. P., and Goudet, C. (2011). Dimers and beyond: the functional puzzles of class C GPCRs. Pharmacol. Ther. 130, 9–25. doi: 10.1016/j.pharmthera.2011.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Kobayashi, S., Conforti, L., and Millhorn, D. E. (2000). Gene expression and function of adenosine A(2A) receptor in the rat carotid body. Am. J. Physiol. Lung Cell. Mol. Physiol. 279, L273–L282. doi: 10.1152/ajplung.2000.279.2.L273

PubMed Abstract | CrossRef Full Text | Google Scholar

Koerner, P., Hesslinger, C., Schaefermeyer, A., Prinz, C., and Gratzl, M. (2004). Evidence for histamine as a transmitter in rat carotid body sensor cells. J. Neurochem. 91, 493–500. doi: 10.1111/j.14714159.2004.02740.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Köse, M. (2017). GPCRs and EGFR – cross-talk of membrane receptors in cancer. Bioorg. Med. Chem. Lett. 27, 3611–3620. doi: 10.1016/j.bmcl.2017.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Kroeger, K. M., Pfleger, K. D. G., and Eidne, K. A. (2003). G-protein coupled receptor oligomerization in neuroendocrine pathways. Front. Neuroendocrinol. 24, 254–278. doi: 10.1016/j.yfrne.2003.10.002

CrossRef Full Text | Google Scholar

Kuszak, A. J., Pitchiaya, S., Anand, J. P., Mosberg, H. I., Walter, N. G., and Sunahara, R. K. (2009). Purification and functional reconstitution of monomeric μ-opioid receptors: allosteric modulation of agonist binding by Gi2. J. Biol. Chem. 284, 26732–26741. doi: 10.1074/jbc.M109.026922

PubMed Abstract | CrossRef Full Text | Google Scholar

Lam, S. Y., Liu, Y., Ng, K. M., Liong, E. C., Tipoe, G. L., Leung, P. S., et al. (2014). Upregulation of a local renin-angiotensin system in the rat carotid body during chronic intermittent hypoxia. Exp. Physiol. 99, 220–231. doi: 10.1113/expphysiol.2013.074591

PubMed Abstract | CrossRef Full Text | Google Scholar

Lambert, N. A., and Javitch, J. A. (2014). CrossTalk opposing view: weighing the evidence for class A GPCR dimers, the jury is still out. J. Physiol. 592, 2443–2445. doi: 10.1113/jphysiol.2014.272997

PubMed Abstract | CrossRef Full Text | Google Scholar

Lazarov, N. E., Reindl, S., Fischer, F., and Gratzl, M. (2009). Histaminergic and dopaminergic traits in the human carotid body. Respir. Physiol. Neurobiol. 165, 131–136. doi: 10.1016/j.resp.2008.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, F. J., Xue, S., Pei, L., Vukusic, B., Chéry, N., Wang, Y., et al. (2002). Dual regulation of NMDA receptor functions by direct protein-protein interactions with the dopamine D1 receptor. Cell 111, 219–230. doi: 10.1016/S0092-8674(02)00962-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. P., So, C. H., Rashid, A. J., Varghese, G., Cheng, R., Lança, A. J., et al. (2004). Dopamine D1 and D2 receptor co-activation generates a novel phospholipase C-mediated calcium signal. J. Biol. Chem. 279, 35671–35678. doi: 10.1074/jbc.M401923200

PubMed Abstract | CrossRef Full Text | Google Scholar

Leonard, E. M., and Nurse, C. A. (2017). “Evidence for dopaminergic inhibitory cell-cell interactions in the rat carotid body,” in XXth Meeting of the International Society of Arterial Chemoreception (ISAC), (Baltimore, MD).

Google Scholar

Leonard, E. M., Salman, S., and Nurse, C. A. (2018). Sensory processing and integration at the carotid body tripartite synapse: neurotransmitter functions and effects of chronic hypoxia. Front. Physiol. 9:225. doi: 10.3389/fphys.2018.00225

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y. C., Liu, G., Hu, J. L., Gao, W. J., and Huang, Y. Q. (2010). Dopamine D(1) receptor-mediated enhancement of NMDA receptor trafficking requires rapid PKC-dependent synaptic insertion in the prefrontal neurons. J. Neurochem. 114, 62–73. doi: 10.1111/j.14714159.2010.06720.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, J., Wei, X., Zhao, C., Hu, S., Duan, J., Ju, G., et al. (2011). 5-HT induces enhanced phrenic nerve activity via 5-HT2Areceptor/PKC mechanism in anesthetized rats. Eur. J. Pharmacol. 657, 67–75. doi: 10.1016/j.ejphar.2011.01.048

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Ji, E. S., Xiang, S., Tamisier, R., Tong, J., Huang, J., et al. (2009). Exposure to cyclic intermittent hypoxia increases expression of functional NMDA receptors in the rat carotid body. J. Appl. Physiol. 106, 259–267. doi: 10.1152/japplphysiol.90626.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Livermore, S., and Nurse, C. A. (2013). Enhanced adenosine A2b receptor signaling facilitates stimulus-induced catecholamine secretion in chronically hypoxic carotid body type I cells. Am. J. Physiol. Cell Physiol. 305, C739–C750. doi: 10.1152/ajpcell.00137

PubMed Abstract | CrossRef Full Text | Google Scholar

Lohse, M. J. (2010). Dimerization in GPCR mobility and signaling. Curr. Opin. Pharmacol. 10, 53–58. doi: 10.1016/j.coph.2009.10.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopez, A., and Salome, L. (2009). Membrane functional organisation and dynamic of mu-opioid receptors. Cell. Mol. Life Sci. 66, 2093–2108. doi: 10.1007/s00018-009-0008-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Lukasiewicz, S., Blasiak, E., Faron-Gorecka, A., Polit, A., Tworzydlo, M., Gorecki, A., et al. (2007). Fluorescence studies of homooligomerization of adenosine A2A and serotonin 5-HT1A receptors reveal the specificity of receptor interactions in the plasma membrane. Pharmacol. Rep. 59, 379–392.

PubMed Abstract | Google Scholar

Lukasiewicz, S., Polit, A., Kedracka-Krok, S., Wedzony, K., Mackowiak, M., and Dziedzicka-Wasylewska, M. (2010). Hetero-dimerization of serotonin 5-HT(2A) and dopamine D(2) receptors. Biochim. Biophys. Acta 1803, 1347–1358. doi: 10.1016/j.bbamcr.2010.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Mackie, K. (2005). Cannabinoid receptor homo- and heterodimerization. Life Sci. 77, 1667–1673. doi: 10.1016/j.lfs.2005.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Marcellino, D., Carriba, P., Filip, M., Borgkvist, A., Frankowska, M., Bellido, I., et al. (2008). Antagonistic cannabinoid CB1/dopamine D2receptor interactions in striatal CB1/D2heteromers. A combined neurochemical and behavioral analysis. Neuropharmacology 54, 815–823. doi: 10.1016/j.neuropharm.2007.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazzatenta, A., Marconi, G. D., Macchi, V., Porzionato, A., Cataldi, A., Di Giulio, C., et al. (2016). Coexpression of galanin and nestin in the chemoreceptor cells of the human carotid body. Adv. Exp. Med. Biol. 885, 77–82. doi: 10.1007/5584_2015_189

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazzatenta, A., Marconi, G. D., Zara, S., Cataldi, A., Porzionato, A., and Di Giulio, C. (2014). In the carotid body, galanin is a signal for neurogenesis in young, and for neurodegeneration in the old and in drug-addicted subjects. Front. Physiol. 5:427. doi: 10.3389/fphys.2014.00427

PubMed Abstract | CrossRef Full Text | Google Scholar

McLemore, G. L. (2004). Cannabinoid receptor expression in peripheral arterial chemoreceptors during postnatal development. J. Appl. Physiol. 97, 1486–1495. doi: 10.1152/japplphysiol.00378.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Möller, D., Salama, I., Kling, R. C., Hubner, H., and Gmeiner, P. (2015). 1,4-Disubstituted aromatic piperazines with high 5-HT2A/D2 selectivity: quantitative structure-selectivity investigations, docking, synthesis and biological evaluation. Bioorg. Med. Chem. 23, 6195–6209. doi: 10.1016/j.bmc.2015.07.050

PubMed Abstract | CrossRef Full Text | Google Scholar

Moreno, E., Hoffmann, H., Gonzalez-Sepulveda, M., Navarro, G., Casado, V., Cortes, A., et al. (2011). Dopamine D1-histamine H3 receptor heteromers provide a selective link to MAPK signaling in GABAergic neurons of the direct striatal pathway. J. Biol. Chem. 286, 5846–5854. doi: 10.1074/jbc.M110.161489

PubMed Abstract | CrossRef Full Text | Google Scholar

Murali, S., Zhang, M., and Nurse, C. A. (2014). Angiotensin II mobilizes intracellular calcium and activates pannexin-1 channels in rat carotid body type II cells via AT1 receptors. J. Physiol. 592, 4747–4762. doi: 10.1113/jphysiol.2014.279299

PubMed Abstract | CrossRef Full Text | Google Scholar

Murali, S., Zhang, M., and Nurse, C. A. (2015). Paracrine signaling in glial-like type II cells of the rat carotid body. Adv. Exp. Med. Biol. 860, 41–47. doi: 10.1007/978-3-319-18440-1_5

PubMed Abstract | CrossRef Full Text | Google Scholar

Nai, Q., Li, S., Wang, S. H., Liu, J., Lee, F. J., Frankland, P. W., et al. (2010). Uncoupling the D1-N-methyl-D-aspartate (NMDA) receptor complex promotes NMDA-dependent long-term potentiation and working memory. Biol. Psychiatry 67, 246–254. doi: 10.1016/j.biopsych.2009.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakata, H., Suzuki, T., Namba, K., and Oyanagi, K. (2010). Dimerization of G protein-coupled purinergic receptors: increasing the diversity of purinergic receptor signal responses and receptor functions. J. Recept. Signal Transduct. Res. 30, 337–346. doi: 10.3109/10799893.2010.509729

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakata, H., Yoshioka, K., Kamiya, T., Tsuga, H., and Oyanagi, K. (2005). Functions of heteromeric association between adenosine and P2Y receptors. J. Mol. Neurosci 26, 233–238. doi: 10.1385/JMN

CrossRef Full Text | Google Scholar

Navarro, G., Aguinaga, D., Moreno, E., Hradsky, J., Reddy, P. P., Cortés, A., et al. (2014). Intracellular calcium levels determine differential modulation of allosteric interactions within G protein-coupled receptor heteromers. Chem. Biol. 21, 1546–1566. doi: 10.1016/j.chembiol.2014.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Nurse, C. A. (2005). Neurotransmission and neuromodulation in the chemosensory carotid body. Auton. Neurosci. 120, 1–9. doi: 10.1016/j.autneu.2005.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Nurse, C. A. (2014). Synaptic and paracrine mechanisms at carotid body arterial chemoreceptors. J. Physiol. 592, 3419–3426. doi: 10.1113/jphysiol.2013.269829

CrossRef Full Text | Google Scholar

Nurse, C. A., Leonard, E. M., and Salman, S. (2018). The role of glial-like type II cells as paracrine modulators of carotid body chemoreception. Physiol. Genomics. 50, 255–262. doi: 10.1152/physiolgenomics.00142.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Nurse, C. A., and Vollmer, C. (1997). Role of basic FGF and oxygen in control of proliferation, survival, and neuronal differentiation in carotid body chromaffin cells. Dev. Biol. 184, 197–206. doi: 10.1006/dbio.1997.8539

PubMed Abstract | CrossRef Full Text | Google Scholar

Oomori, Y., Ishikawa, K., Satho, Y., Matsuda, M., and Ono, K. (1991). Neuropeptide-Y-immunoreactive chief cells in the carotid body of young rats. Acta Anat. 140, 120–123. doi: 10.1159/000147046

PubMed Abstract | CrossRef Full Text | Google Scholar

Oomori, Y., Murabayashi, H., Ishikawa, K., Miyakawa, K., Nakaya, K., and Tanaka, H. (2002). Neuropeptide Y- and catecholamine-synthesizing enzymes: immunoreactivities in the rat carotid body during postnatal development. Anat. Embryol. 206, 37–47. doi: 10.1007/s00429-002-0275-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Ortega-Sáenz, P., Villadiego, J., Pardal, R., Toledo-Aral, J. J., and Lopez-Barneo, J. (2015). Neurotrophic properties, chemosensory responses and neurogenic niche of the human carotid body. Adv. Exp. Med. Biol. 860, 139–152. doi: 10.1007/978-3-319-18440-1_16

PubMed Abstract | CrossRef Full Text | Google Scholar

Paciga, M., and Nurse, C. A. (2001). Basic FGF localization in rat carotid body: paracrine role in O2-chemoreceptor survival. Neuroreport 12, 3287–3291. doi: 10.1097/00001756-200110290-00028

PubMed Abstract | CrossRef Full Text | Google Scholar

Palczewski, K. (2010). Oligomeric forms of G protein-coupled receptors (GPCRs). Trends Biochem. Sci. 35, 595–600. doi: 10.1016/j.tibs.2010.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Pardal, R., Ortega-Sàenz, P., Duràn, R., and Lòpez-Barneo, J. (2007). Glia-like stem cells sustain physiologic neurogenesis in the adult mammalian carotid body. Cell 131, 364–377. doi: 10.1016/j.cell.2007.07.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, P. S. H., Filipek, S., Wells, J. W., and Palczewski, K. (2004). Oligomerization of G protein-coupled receptors: past, present, and future. Biochemistry 43, 15643–15656. doi: 10.1021/bi047907k

PubMed Abstract | CrossRef Full Text | Google Scholar

Pawar, A., Nanduri, J., Yuan, G., Khan, S. A., Wang, N., Kumar, G. K., et al. (2009). Reactive oxygen species-dependent endothelin signaling is required for augmented hypoxic sensory response of the neonatal carotid body by intermittent hypoxia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 296, R735–R742. doi: 10.1152/ajpregu.90490.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, Y. J., Nanduri, J., Raghuraman, G., Wang, N., Kumar, G. K., and Prabhakar, N. R. (2013). Role of oxidative stress-induced endothelin-converting enzyme activity in the alteration of carotid body function by chronic intermittent hypoxia. Exp. Physiol. 98, 1620–1630. doi: 10.1113/expphysiol.2013.073700

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, Y. J., Nanduri, J., Yuan, G., Wang, N., Deneris, E., Pendyala, S., et al. (2009). NADPH oxidase is required for the sensory plasticity of the carotid body by chronic intermittent hypoxia. J. Neurosci. 29, 4903–4910. doi: 10.1523/JNEUROSCI.4768-08.2009

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, Y. J., Overholt, J. L., Kline, D., Kumar, G. K., and Prabhakar, N. R. (2003). Induction of sensory long-term facilitation in the carotid body by intermittent hypoxia: implications for recurrent apneas. Proc. Natl. Acad. Sci. U.S.A. 100, 10073–10078. doi: 10.1073/pnas.1734109100

PubMed Abstract | CrossRef Full Text | Google Scholar

Piskuric, N. A., and Nurse, C. A. (2013). Expanding role of ATP as a versatile messenger at carotid and aortic body chemoreceptors. J. Physiol. 591, 415–422. doi: 10.1113/jphysiol.2012.234377

PubMed Abstract | CrossRef Full Text | Google Scholar

Platero-Luengo, A., González-Granero, S., Durán, R., Díaz-Castro, B., Piruat, J. I., García-Verdugo, J. M., et al. (2014). An O2-sensitive glomus cell-stem cell synapse induces carotid body growth in chronic hypoxia. Cell 156, 291–303. doi: 10.1016/j.cell.2013.12.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Pontier, S. M., Lahaie, N., Ginham, R., St-Gelais, F., Bonin, H., Bell, D. J., et al. (2006). Coordinated action of NSF and PKC regulates GABAB receptor signaling efficacy. EMBO J. 25, 2698–2709. doi: 10.1038/sj.emboj.7601157

PubMed Abstract | CrossRef Full Text | Google Scholar

Porzionato, A., Macchi, V., Amagliani, A., Castagliuolo, I., Parenti, A., and De Caro, R. (2009). Neurotensin receptor 1 immunoreactivity in the peripheral ganglia and carotid body. Eur. J. Histochem. 53, 135–142. doi: 10.4081/ejh.2009.e16

PubMed Abstract | CrossRef Full Text | Google Scholar

Porzionato, A., Macchi, V., Barzon, L., Masi, G., Belloni, A., Parenti, A., et al. (2010). Expression and distribution of galanin receptor subtypes in the rat carotid body. Mol. Med. Rep. 3, 37–41. doi: 10.3892/mmr_00000215

PubMed Abstract | CrossRef Full Text | Google Scholar

Porzionato, A., Macchi, V., Parenti, A., Matturri, L., and De Caro, R. (2008a). Peripheral chemoreceptors: postnatal development and cytochemical findings in Sudden Infant Death Syndrome. Histol. Histopathol. 23, 351–365. doi: 10.14670/HH-23.351

PubMed Abstract | CrossRef Full Text | Google Scholar

Porzionato, A., Macchi, V., Parenti, A., and De Caro, R. (2008b). Trophic factors in the carotid body. Int. Rev. Cell Mol. Biol. 269, 1–58. doi: 10.1016/S1937-6448(08)01001-0

CrossRef Full Text | Google Scholar

Porzionato, A., Macchi, V., Stecco, C., and De Caro, R. (2018). The carotid sinus nerve – structure, function and clinical implications. Anat. Rec. doi: 10.1002/ar.23829 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Prabhakar, N. R. (2011). Sensory plasticity of the carotid body: role of reactive oxygen species and physiological significance. Respir. Physiol. Neurobiol. 178, 375–380. doi: 10.1016/j.resp.2011.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Rashid, A. J., So, C. H., Kong, M. M. C., Furtak, T., El-Ghundi, M., Cheng, R., et al. (2007). D1-D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc. Natl. Acad. Sci. U.S.A. 104, 654–659. doi: 10.1073/pnas.0604049104

PubMed Abstract | CrossRef Full Text | Google Scholar

Rey, S., Corthorn, J., Chacón, C., and Iturriaga, R. (2007). Expression and immunolocalization of endothelin peptides and its receptors, ETA and ETB, in the carotid body exposed to chronic intermittent hypoxia. J. Histochem. Cytochem. 55, 167–174. doi: 10.1369/jhc.6A7079.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Rey, S., Del Rio, R., Alcayaga, J., and Iturriaga, R. (2004). Chronic intermittent hypoxia enhances cat chemosensory and ventilatory responses to hypoxia. J. Physiol. 560, 577–586. doi: 10.1113/jphysiol.2004.072033

PubMed Abstract | CrossRef Full Text | Google Scholar

Ricker, E. M., Pye, R. L., Barr, B. L., and Wyatt, C. N. (2015). Selective mu and kappa opioid agonists inhibit voltage-gated Ca2+ entry in isolated neonatal rat carotid body type I cells. Adv. Exp. Med. Biol. 860, 49–54. doi: 10.1007/978-3-319-18440-1_6

PubMed Abstract | CrossRef Full Text | Google Scholar

Rios, C. D., Jordan, B. A., Gomes, I., and Devi, L. A. (2001). G-protein-coupled receptor dimerization: modulation of receptor function. Pharmacol. Ther. 92, 71–87. doi: 10.1016/S0163-7258(01)00160-7

CrossRef Full Text | Google Scholar

Rocher, A., Gonzalez, C., and Almaraz, L. (1999). Adenosine inhibits L-type Ca2+ current and catecholamine release in the rabbit carotid body chemoreceptor cells. Eur. J. Neurosci. 11, 673–681. doi: 10.1046/j.1460-9568.1999.00470.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy, A., Mandadi, S., Fiamma, M.-N., Rodikova, E., Ferguson, E. V., Whelan, P. J., et al. (2012). Anandamide modulates carotid sinus nerve afferent activity via TRPV1 receptors increasing responses to heat. J. Appl. Physiol. 112, 212–224. doi: 10.1152/japplphysiol.01303.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, D. K., Sengupta, A., Zhang, C., Boyadjieva, N., and Murugan, S. (2012). Opiate antagonist prevents μ- and δ-opiate receptor dimerization to facilitate ability of agonist to control ethanol-altered natural killer cell functions and mammary tumor growth. J. Biol. Chem. 287, 16734–16747. doi: 10.1074/jbc.M112.347583

PubMed Abstract | CrossRef Full Text | Google Scholar

Satake, H., Matsubara, S., Aoyama, M., Kawada, T., and Sakai, T. (2013). GPCR heterodimerization in the reproductive system: functional regulation and implication for biodiversity. Front. Endocrinol. (Lausanne). 4:100. doi: 10.3389/fendo.2013.00100

PubMed Abstract | CrossRef Full Text | Google Scholar

Schicker, K., Hussl, S., Chandaka, G. K., Kosenburger, K., Yang, J. W., Waldhoer, M., et al. (2009). A membrane network of receptors and enzymes for adenine nucleotides and nucleosides. Biochim. Biophys. Acta 1793, 325–334. doi: 10.1016/j.bbamcr.2008.09.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Schiffmann, S. N., Dassesse, D., d’Alcantara, P., Ledent, C., Swillens, S., and Zoli, M. (2003). A2A receptor and striatal cellular functions: regulation of gene expression, currents, and synaptic transmission. Neurology 61, S24–S29. doi: 10.1212/01.WNL.0000095207.66853.0D

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlenker, E. H. (2008). In hamsters the D1 receptor antagonist SCH 23390 depresses ventilation during hypoxia. Brain Res. 1187, 146–153. doi: 10.1016/j.brainres.2007.10.058

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlenker, E. H., and Schultz, H. D. (2011). Hypothyroidism attenuates SCH 23390-mediated depression of breathing and decreases D1 receptor expression in carotid bodies, PVN and striatum of hamsters. Brain Res. 1401, 40–51. doi: 10.1016/j.brainres.2011.05.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Schonenbach, N. S., Hussain, S., and O’Malley, M. A. (2015). Structure and function of G protein-coupled receptor oligomers: implications for drug discovery. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 7, 408–427. doi: 10.1002/wnan.1319

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, J., Zhang, L., Song, W., Meng, T., Wang, X., Chen, L., et al. (2013). Design, synthesis and biological evaluation of bivalent ligands against A1–D1 receptor heteromers. Acta Pharmacol. Sin. 34, 441–452. doi: 10.1038/aps.2012.151

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirahata, M., Hirasawa, S., Okumura, M., Mendoza, J. A., Okumura, A., Balbir, A., et al. (2004). Identification of M1 and M2 muscarinic acetylcholine receptors in the cat carotid body chemosensory system. Neuroscience 128, 635–644. doi: 10.1016/j.neuroscience.2004.06.068

PubMed Abstract | CrossRef Full Text | Google Scholar

Skieterska, K., Duchou, J., Lintermans, B., and Van Craenenbroeck, K. (2013). Detection of G protein-coupled receptor (GPCR) dimerization by coimmunoprecipitation. Methods Cell Biol. 117, 323–340. doi: 10.1016/B978-0-12-408143-7.00017-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Stockert, J. A., and Devi, L. A. (2015). Advancements in therapeutically targeting orphan GPCRs. Front. Pharmacol. 6:100. doi: 10.3389/fphar.2015.00100

PubMed Abstract | CrossRef Full Text | Google Scholar

Stockwell, J., Chen, Z., Niazi, M., Nosib, S., and Cayabyab, F. S. (2016). Protein phosphatase role in adenosine A1 receptor-induced AMPA receptor trafficking and rat hippocampal neuronal damage in hypoxia/reperfusion injury. Neuropharmacology 102, 254–265. doi: 10.1016/j.neuropharm.2015.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Tao, Y., Yu, C., Wang, W., Hou, Y., Xu, X., Chi, Z., et al. (2017). Heteromers of μ opioid and dopamine D1 receptors modulate opioid-induced locomotor sensitization in a dopamine-independent manner. Br. J. Pharmacol. 174, 2842–2861. doi: 10.1111/bph.13908

PubMed Abstract | CrossRef Full Text | Google Scholar

Terrillon, S., and Bouvier, M. (2004). Roles of G-protein-coupled receptor dimerization. EMBO Rep. 5, 30–34. doi: 10.1038/sj.embor.7400052

PubMed Abstract | CrossRef Full Text | Google Scholar

Thompson, C. M., Troche, K., Jordan, H. L., Barr, B. L., and Wyatt, C. N. (2010). Evidence for functional, inhibitory, histamine H3 receptors in rat carotid body type I cells. Neurosci. Lett. 471, 15–19. doi: 10.1016/j.neulet.2009.12.077

PubMed Abstract | CrossRef Full Text | Google Scholar

Tjong, Y. W., Chen, Y., Liong, E. C., Ip, S. F., Tipoe, G. L., and Fung, M. L. (2004). Melatonin attenuates rat carotid chemoreceptor response to hypercapnic acidosis. J. Pineal Res. 36, 49–57. doi: 10.1046/j.1600-079X.2003.00094.x

CrossRef Full Text | Google Scholar

Trifilieff, P., Rives, M.-L., Urizar, E., Piskorowski, R. A., Vishwasrao, H. D., Castrillon, J., et al. (2011). Detection of antigen interactions ex vivo by proximity ligation assay: endogenous dopamine D2-adenosine A2A receptor complexes in the striatum. Biotechniques 51, 111–118. doi: 10.2144/000113719

PubMed Abstract | CrossRef Full Text | Google Scholar

Tse, A., Yan, L., Lee, A. K., and Tse, F. W. (2012). Autocrine and paracrine actions of ATP in rat carotid body. Can. J. Physiol. Pharmacol. 90, 705–711. doi: 10.1139/y2012-054

PubMed Abstract | CrossRef Full Text | Google Scholar

Tuteja, N. (2009). Signaling through G protein coupled receptors. Plant Signal. Behav. 4, 942–947. doi: 10.4161/psb.4.10.9530

CrossRef Full Text | Google Scholar

Wakai, J., Takayama, A., Yokoyama, T., Nakamuta, N., Kusakabe, T., and Yamamoto, Y. (2015). Immunohistochemical localization of dopamine D2 receptor in the rat carotid body. Acta Histochem. 117, 784–789. doi: 10.1016/j.acthis.2015.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, D., Sun, X., Bohn, L., and Sadee, W. (2005). Opioid receptor homo-and heterodimerization in living cells by quantitative bioluminescence resonance energy transfer. Mol. Pharmacol. 67, 2173–2184. doi: 10.1124/mol.104.010272.cept

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Z. Y., and Bisgard, G. E. (2002). Chronic hypoxia-induced morphological and neurochemical changes in the carotid body. Microsc. Res. Tech. 59, 168–177. doi: 10.1002/jemt.10191

PubMed Abstract | CrossRef Full Text | Google Scholar

Whorton, M. R., Bokoch, M. P., Rasmussen, S. G., Huang, B., Zare, R. N., Kobilka, B., et al. (2007). A monomeric G protein-coupled receptor isolated in a high-density lipoprotein particle efficiently activates its G protein. Proc. Natl. Acad. Sci. U.S.A. 104, 7682–7687. doi: 10.1073/pnas.0611448104

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, H., Hu, L., and Li, G. (2010). SH-SY5Y human neuroblastoma cell line: in vitro cell model of dopaminergic neurons in Parkinson’s disease. Chin. Med. J. (Engl). 123, 1086–1092.

Google Scholar

Xu, J., Tse, F. W., and Tse, A. (2003). ATP triggers intracellular Ca2+ release in type II cells of the rat carotid body. J. Physiol. 549, 739–747. doi: 10.1113/jphysiol.2003.039735

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, J., Xu, F., Tse, F. W., and Tse, A. (2005). ATP inhibits the hypoxia response in type I cells of rat carotid bodies. J. Neurochem. 92, 1419–1430. doi: 10.1111/j.1471-4159.2004.02978.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yekkirala, A. S., Kalyuzhny, A. E., and Portoghese, P. S. (2013). An immunocytochemical-derived correlate for evaluating the bridging of heteromeric mu-delta opioid protomers by bivalent ligands. ACS Chem. Biol. 8, 1412–1416. doi: 10.1021/cb400113d

PubMed Abstract | CrossRef Full Text | Google Scholar

Yokoyama, T., Nakamuta, N., Kusakabe, T., and Yamamoto, Y. (2015). Serotonin-mediated modulation of hypoxia-induced intracellular calcium responses in glomus cells isolated from rat carotid body. Neurosci. Lett. 597, 149–153. doi: 10.1016/j.neulet.2015.04.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshioka, K., Saitoh, O., and Nakata, H. (2001). Heteromeric association creates a P2Y-like adenosine receptor. Proc. Natl. Acad. Sci. U.S.A. 98, 7617–7622. doi: 10.1073/pnas.121587098

PubMed Abstract | CrossRef Full Text | Google Scholar

Zara, S., Pokorski, M., Cataldi, A., Porzionato, A., De Caro, R., Antosiewicz, J., et al. (2013). Development and aging are oxygen-dependent and correlate with VEGF and NOS along life span. Adv. Exp. Med. Biol. 756, 223–228. doi: 10.1007/978-94-007-4549-0_28

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, C., Wang, Z., Asico, L. D., Hopfer, U., Eisner, G. M., Felder, R. A., et al. (2005). Aberrant ETB receptor regulation of AT1 receptors in immortalized renal proximal tubule cells of spontaneously hypertensive rats. Kidney Int. 68, 623–631. doi: 10.1111/j.1523-1755.2005.00440.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, M., Fearon, I. M., Zhong, H., and Nurse, C. A. (2003). Presynaptic modulation of rat arterial chemoreceptor function by 5-HT: role of K+ channel inhibition via protein kinase C. J. Physiol. 551, 825–842. doi: 10.1113/jphysiol.2002.038489

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, M., Vollmer, C., and Nurse, C. A. (2017). Adenosine and dopamine oppositely modulate a hyperpolarization-activated current Ih in chemosensory neurons of the rat carotid body in co-culture. J. Physiol. doi: 10.1113/JP274743 [Epub ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhong, H., and Nurse, C. (1995). Basic fibroblast growth factor regulates ionic currents and excitability of fetal rat carotid body chemoreceptors. Neurosci. Lett. 202, 41–44. doi: 10.1016/0304-3940(95)12200-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Zidar, D. A., Violin, J. D., Whalen, E. J., and Lefkowitz, R. J. (2009). Selective engagement of G protein coupled receptor kinases (GRKs) encodes distinct functions of biased ligands. Proc. Natl. Acad. Sci. U.S.A. 106, 9649–9654. doi: 10.1073/pnas.0904361106

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: carotid body, GPCRs, heteromers, receptor mosaics, hypoxia, adenosine, dopamine, plasticity

Citation: Porzionato A, Stocco E, Guidolin D, Agnati L, Macchi V and De Caro R (2018) Receptor–Receptor Interactions of G Protein-Coupled Receptors in the Carotid Body: A Working Hypothesis. Front. Physiol. 9:697. doi: 10.3389/fphys.2018.00697

Received: 07 April 2018; Accepted: 18 May 2018;
Published: 07 June 2018.

Edited by:

Rodrigo Iturriaga, Pontificia Universidad Católica de Chile, Chile

Reviewed by:

Ricardo Fernandez, University of Los Lagos, Chile
Joana F. Sacramento, Universidade Nova de Lisboa, Portugal

Copyright © 2018 Porzionato, Stocco, Guidolin, Agnati, Macchi and De Caro. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Andrea Porzionato, andrea.porzionato@unipd.it

These authors have contributed equally to this work.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.