key: cord-0037793-fw5tcnrx authors: Thiriet, Marc title: G-Protein-Coupled Receptors date: 2011-12-08 journal: Signaling at the Cell Surface in the Circulatory and Ventilatory Systems DOI: 10.1007/978-1-4614-1991-4_7 sha: 2eef409a67f16b6ef6aa2a5afe62e9c3ccd09096 doc_id: 37793 cord_uid: fw5tcnrx Receptors coupled to heterotrimeric guanine nucleotide-binding proteins, the socalled G-protein-coupled receptors (GPCR), represent the largest set of plasmalemmal receptors Agonist binding does not always cause important conformational changes. Moreover, agonist subtypes can be classified according to their effect on the receptor structure [762] . Some ligands stabilize a given receptor conformation (conformationally selective ligands). Others modify the receptor structure among multiple receptor conformations and shift the dynamical equilibrium. Human adenosine A 2A receptor and β-adrenergic receptors possess binding sites that can bind specifically to agonists rather than antagonists and inverse agonists [763] . Inverse agonists can bind to a given GPCR to prevent the conformational change that primes its activation. An intermediate conformation between the inactive and active states is actually characterized by an occlusion of the G-protein-binding site. In addition to neutral antagonists and inverse agonists, full and partial agonists prime various rearrangements of the receptor structure that initiate signaling via Gproteins and/or arrestins (Sect. 7.11). 1 Full agonists are capable of maximal receptor stimulation, whereas partial agonists are unable to elicit full activity even at high concentration. A given conformation stabilized by a ligand thus determines the efficacy toward a specific pathway. In summary, a ligand can operate as: (1) a full agonist that is capable of activating the receptor, thereby causing an a priori maximal response; (2) partial agonist that does not activate its cognate receptors thoroughly, hence provoking partial response; (3) inverse agonist that reduces the activity of its receptor by inhibiting their constitutive activity; and (4) antagonist that binds to the receptor, blocks action of agonists, as it prevents access to the receptor by its binding. In addition, many GPCRs can stimulate multiple signaling systems, and a given ligand can excite different pathways with different relative efficiency up to opposite activity. For example, agonists of β2-adrenoceptors for the arrestin-MAPK pathway act as antagonists for the Gs-cAMP-PKA axis. Alternative splicing diversifies the receptorome and augments the number of GPCR types encountered on cell surfaces, hence heterogeneity in GPCR signaling. In humans, airway smooth muscle cells are endowed with multiple types (>350) of G-protein-coupled receptors. More than 190 GPCRs have, on average, 5 different receptor isoforms that can result from various ways of alternative splicing of pre-mRNAs [764] (Vol. 1 -Chap. 5. Protein Synthesis). 7.1 Introduction Table 7 .1. Types of subunits of guanine nucleotide-binding (G) proteins and triggered pathways (Source: [765] ; ACase: adenylate cyclase; cAMP: cyclic adenosine monophosphate; DAG: diacylglycerol; cGMP: cyclic guanosine monophosphate; GEF: guanine nucleotideexchange factor; IP 3 : inositol (1,4,5)-trisphosphate; PDE: phosphodiesterase; PI3K: phosphatidylinositol 3-kinase; PIP 3 : phosphatidylinositol triphosphate; PKC: protein kinase C; PKG: protein kinase G; PLC: phospholipase C; −→: inhibition). Targets and signaling axis Table 7 .4. Ligands of G-protein-coupled receptors: (Part 1) amino acids, dicarboxylic acids, and biogenic amines (Source: [767] ; GABA: γ-aminobutyric acid). Amino acids L Arginine, L lysine GPRc6a aGq/11 Glutamate mGlu 1,5 Gq/11 mGlu 2, 3, 4, 6, 7, 8 Gi The vasculature can experience an organ-specific development. In particular, the specialized vasculature of the central nervous system possesses a strong limitation in molecular permeability through the strongly polarized, 4 tightly sealed endothelial cells, extensive pericyte coverage, and reciprocal interactions with neurons and glial cells. Moreover, it acts as a neural stem cell niche. The blood-brain barrier is able to insulate the central nervous system from substances that can be tolerated by peripheral organs. The pro-angiogenic, endothelial, adhesion G-protein-coupled receptor GPR124 5 is involved in the development of endothelial cells and pericytes of the 4. Molecular carriers are heterogeneously distributed between the luminal and abluminal edges of the plasma membrane. 5. A.k.a. tumor endothelial marker-5 (TEM5). During mouse embryogenesis, GPR124 is expressed in both endothelial cells and pericytes, mainly in the neural tube, and, to a lesser extent, the heart, liver, and kidney, as well as the epithelium of embryonic lung and esophagus Table 7 .5. Ligands of G-protein-coupled receptors: (Part 2) ions, nucleotides, and nucleosides (Sources: [767, 769, 770] ). Proton-sensing G-protein-coupled receptors sense acidic pH. The GPR4 receptor (or G2 accumulation protein [G2A]) has a low affinity for lysophosphatidylcholine (LPC) and high affinity for sphingosylphosphorylcholine (SPC) . On the other hand, GPR132 has a high affinity for LPC and low affinity for SPC. Receptors GPR65 and GPR68 are also called T-cell death-associated gene-8 protein (TDAG8) and ovarian cancer GPCR OGR1 as well as SPC receptor SPC 1 , respectively. The GPR4 receptor is involved in angiogenesis primed by SPC in endothelial cells via phosphatidylinositol 3-kinase, protein kinase-B, and vascular endothelial growth factor receptor VEGFR2. Receptor cerebral vasculature, particularly during CNS-specific angiogenesis that forms the perineural, then periventricular vascular plexi in the forebrain, ventral neural tube, and spinal cord [772, 773] . 6 During adulthood, the brain endothelium remains sensitive to GPR124 receptor. The recruitment of blood-circulating phagocytes to sites of inflammation and infection begins with activation and interactions of cell adhesion receptors on both leukocytes and endothelial cells. Phagocyte activation is regulated partly by celland mesenchyme. In adult mice, it is exclusively produced in the vasculature with more developed pericyte coating, i.e., in the brain and kidney, pancreas, and corpus luteum [772] . The GPR124 receptor is required for invasion and migration of blood vessels into the neuroepithelium, establishment of the blood-brain barrier, and expansion of the cerebral cortex [773] . 6. Deletion of GPR124 causes angiogenesis arrest; overexpression hyperproliferative vascular malformations [772] . The GRP124 receptor regulates CDC42-dependent angiogenic migration and sprouting. Mutations of genes of the Wnt-βCtn and VEGF-Nrp1 pathways, of the helix-loop-helix transcriptional repressors, the inhibitors of DNA binding ID1 to ID3 required to maintain the timing of neuronal differentiation in the embryo and invasiveness of the vasculature [774] (ID1 is an inhibitor of angiogenesis via thrombospondin-1 [775]; ID2 is a target of retinoblastoma protein sufficient and necessary for the production of VEGF [776] ), and integrin-α V and -β 8 also impair angiogenesis in the central nervous system, but without CNS tropism. Table 7 .6. Ligands of G-protein-coupled receptors: (Part 3) lipids (Source: [767] ; 2AG: 2arachidonyl glycerol; 5oxoETE: 5-oxo (6, 8, 11, 14) -eicosatetraenoic acid; AEA: N arachidonoyl ethanolamine (anandamide); G Tc ; cone-transducin [G T 2 ]; G Tr : rod-transducin [G T 1 ]; LC-, SCFA: long-, short-chain [<6 carbons] fatty acid; OxER: oxoeicosanoid [OXE] receptor; PAF; platelet-activating factor). Agent 11-cis-retinal covalently binds receptors for lightdependent activation. Agent 5oxoETE is produced by oxidation of 5HETE (HETE: hydroxyeicosatrienoic acid) by 5-hydroxyeicosanoid dehydrogenase (5-lipoxygenase pathway). Receptor Gs, Gq/11, Gi TxA2 TP Gq/11, G12/13 Sphingosine-1-phosphate S1P 1 -S1P 5 Gi, Gq/11, G12/13 Sphingosylphosphorylcholine SPC 1 , SPC 2 Gi surface receptors such as adhesion G-protein-coupled receptors. Epidermal growth factor-like module-containing, mucin-like, hormone receptor-like protein EMR2 is synthesized by neutrophils, monocytes, macrophages, and dendritic cells. It binds to chondroitin sulfate on cells and tissue matrix. It regulates the neutrophil response [777] . It potentiates the effects of numerous pro-inflammatory mediators. Upon neutrophil activation, EMR2 is rapidly translocated to membrane ruffles and the leading edge of the migrating cell and its production can rise. Ligation of EMR2 boosts the activation and recruitment of neutrophils, increases neutrophil adhesion may sense renal insults, possibly by primary cilium-mediated mechanotransduction, and trigger a TOR-initiated repair program. 7 Proteins TRPP1 and TRPP2 represent an adhesion G-protein-coupled receptor and a mechanosensitive, calcium-permeable, non-selective cation channel of the TRP channel family located at the primary cilium that causes calcium transients at the cilium, respectively. Protein TRPP1 can interact with the C-terminus of TRPP2, thereby regulating the TRPP2 activity. 8 Protein TRPP1 is a multidomain glycoprotein of 4,303 amino acids. It is made of a large extracellular N-terminal region, a membrane-spanning domain with 11 transmembrane segments, and a cytosolic C-terminal segment (Table 7 .12). The last 7. The primary cilium of renal epithelial cells is a non-motile, mechanosensory extensions of the apical (luminal) plasma membrane that bends in response to urine flow and provokes a transient rise in the intracellular calcium concentration. Polycystin-1 links ciliary mechanosensation to changes in gene transcription via flow-regulated proteolytic cleavage of the TRPP1 cytoplasmic tail, its nuclear translocation, and stimulation of STAT6 transcriptional activity [778] . 8. The cytoplasmic segment serves in the recruitment to the cell surface and interaction with TRPP2 channel. The TRPP1-TRPP2 complex functions as a cation channel. Table 7 .9. Ligands of G-protein-coupled receptors: (Part 4.3) peptides and proteins (Source: [767] ; DOR, KOR, MOR: δ-, κ-, μ-opioid receptor; PAR: peptidase-activated receptor; PrRP: prolactin-releasing peptide; PTH: parathyroid hormone; PTHRP: parathyroid hormone-related protein). N-Termini of peptidases released by proteolytic cleavage serves as ligands. Receptor Gα 6 transmembrane segments are homologous to TRPP2 and voltage-activated calcium channels. In renal tubules, bending of the wetted primary cilium by urine flow activates TRPP1 and TRPP2 proteins. It can couple with and activate several heterotrimeric G protein subtypes, such as Gi, Gq, G12/13, and stimulate Jun N-terminal kinase and AP1 transcription factor [779] . Protein TRPP1 regulates the cell cycle, as it upregulates cyclin-dependent kinase inhibitor CKI1a and activates the JaK-STAT pathway [780] . In addition, TRPP1 binds to and stabilizes regulator of G-protein signaling RGS7. Protein TRPP1, like other adhesion G-protein-coupled receptors, can be cleaved at the GPCR proteolytic site (GPS). 9 Protein TRPP1 actually undergoes an autoproteolytic GPS cleavage to form an extracellular N-terminal fragment and a membranous C-terminal fragment that remain non-covalently associated. Cleavage at GPS is autoproteolytic, but not entirely efficient. Therefore, cleaved and uncleaved adhesion G-protein-coupled receptors coexist. Both full length and cleaved fragment of adhesion G-protein-coupled receptor can be involved in cell signaling. 9 . The GPS domain alone is not sufficient to support the cleavage and requires the adjacent receptor for egg jelly domain [781] . After GPS cleavage, the N-terminal α subunit is anchored to the plasma membrane, as it tethers to transmembrane β subunit or connects to the plasma membrane by itself [782] . In addition, N-and C-termini can be internalized independently. Moreover, α and β subunits of different types of adhesion G-protein-coupled receptors can interact. However, most of the TRPP1 N-terminal fragment (TRPP1 Δ NT ) is tethered to the membrane-bound C-terminal fragment (TRPP1 Δ CT ) in a non-covalent manner (similar to that for long N-terminal class-B GPCR-related 7-transmembrane receptors latrophilin-1 and CD97). Proteolysis at the GPS that occurs at the endoplasmic reticulum may assist in the transfer to the plasma membrane. However, at least for some adhesion G-proteincoupled receptors such as TRPP1, it is not required for an efficient transport to the plasma membrane [782] . In any case, GPS cleavage is mandatory for a normal receptor functioning. The final, mature, plasmalemmal receptor may be a heterodimer or 2 independent proteins [782] . Membrane-bound N-terminus may either be released in the extracellular medium, or remains connected to the cell surface and bind its cognate ligands and then transmits cues. The C-terminal transmembrane segment subunit may act as a classical GPCR; it can reassociate with the liganded N-terminal subunit [782] . Table 7 .11. Ligands of G-protein-coupled receptors: (Part 5) sensory receptors (Source: [767] ; G gust : gustducin; G olf : olfaction G-protein subunit; G Tc and G Tr : cone-and rod-transducin). Visual signals are sensed by opsins that use a photo-isomerization reaction to translate electromagnetic waves into cellular signals. Opsin signaling is based on the conversion of 11-cis-retinal to all-trans-retinal. Light is an multichromatic electromagnetic wave (visible spectrum wavelength 380-780 nm, i.e. 400-790 THz, with a maximum sensitivity ∼ 555 nm). In humans, 4 opsin types exist in addition to rhodopsin. They lodge in different types of cone cells of the retina. They have absorption maxima for yellowish-green, green, and bluish-violet light. Smell sensing is carried out by receptors of the olfactory epithelium that bind odorants (olfactory receptors) and pheromones (vomeronasal receptors). Because of their similar aliases, pheromone receptors V 1 R and V 2 R should not be mistaken for V 1A/1B/2 vasopressin receptors. Receptor Gα Proton-sensing G-protein-coupled receptors pertain to the subclass A15. They sense acidic pH; they are indeed activated when the extracellular pH falls below 6.8 (acidosis, i.e., increased hydrogen ion concentration). 10 They include GPR4, GPR65, 11 GPR68, 12 and GPR132. 13 The latter is an immunoregulatory receptor on macrophages for lysophosphatidylcholine, which is a major phospholipid component of oxidized low-density lipoproteins in atherosclerosis. [783] . The GPR132 receptor resides also on lymphocytes [784] . Activation of GPR132 by lysophosphatidylcholine may increase intracellular calcium concentration, cause receptor internalization, and activate extracellular signal-regulated kinases. The GPR65 receptor on the surface of tumor cells facilitates tumor development by sensing the acidic environment [785] . The superclass of G-protein-coupled receptors contains many hundreds of olfactory receptors to detect a wide variety of olfactory (exogenous) ligands and more than 360 endoGPCRs that are targeted by endogenous (non-olfactory) ligands. The GPCR superclass is constituted by several classes. On the basis of their sequence and structural similarity, ligand interaction, and phylogeny, GPCRs have been originally grouped into A to F or 1 to 5 classes. 14 Phylogenetic analysis of the human repertoire yields the alternative GRAFS classification with 5 families: glutamate (G), rhodopsin (R), adhesion (A), Frizzled-Taste-2 (F), and secretin (S) [788] . The number of elements in each class varies according to literature data, and especially publication date, as new proteins are discovered (Table 7 .13). The class of rhodopsin GPCRs is the largest set with about 672 members in the human genome (∼ 388 olfactory receptors). Its members are often encoded by single exons or genes with a small number of introns. Isomerization of 11-cis-retinal 10. Acidosis is usually detected in blood (acidemia; arterial pH <7.35). 11. A.k.a. T-cell death-associated gene-8 (TDAG8) and psychosine receptor. It is overexpressed in various tumor cell types. 12. A.k.a. ovarian cancer G-protein coupled receptor-1 (OGR1). 13. A.k.a. cell cycle phase G2 accumulation protein (G2A). 14. The families comprise Rhodopsin (class A with more than 270 members), Secretin (at least 15 receptors) and Adhesion (33 members; class B), metabotropic glutamate (class C; 15 receptors), fungal mating pheromone receptors (class D), cAMP receptors (class E), and Frizzled-Smoothened-Taste-2 (class F; 24 members) classes [786, 787] . The very large Rhodopsin class A is subdivided into 19 groups (A1-A19). Class-B GPCRs include both Adhesion and Secretin families. Class-B GPCRs are also called class-2 GPCRs. Taste-2 receptors sense bitter substances. Another classification scheme of GPCRs is based on class 1 Rhodopsin-like, class 2 Secretin-like and Adhesion, class 3 Glutamate receptor-like, class 4 Frizzled-Taste-2, and class 5 Miscellaneous (Others, including unclassified GPCRs). Table 7 .13. Estimated GPCR number per class in humans (Source: [789] ). Rhodopsin consists of the protein moiety opsin that is reversibly, covalently bound to its cofactor retinal. The latter is produced in the retina from vitamin-A. Photo-isomerization of retinal is the primary vision event that creates photosensory signals via retinal interactions with other proteins. into all-trans-retinal by light induces a conformational change in opsin that activates the associated G protein and triggers a signaling cascade. In humans, several opsin types exist in addition to rhodopsin. They reside in different types of cone cells of the retina. They have absorption maxima for yellowish-green (∼ 560 nm), green (∼ 530 nm), and bluish-violet (∼ 426 nm) light (Table 7 .14). The class of adhesion GPCRs that comprises 33 members is the second largest GPCR set in humans. Mammalian adhesion GPCRs are encoded by large, complex genomic structures with several introns that give rise to alternatively spliced variants. The class of secretin GPCRs members have a peptide hormone-binding domain. They likely originates from ancestors to the class of adhesion GPCRs. Class-1 GPCRs, the largest and most diverse class of GPCRs, encompass more than 400 sensory receptor types involved with the detection of taste, odor, or light, as well as more than 250 non-sensory receptor types for transmitters or chemical messengers, in addition to orphan receptors. Class-2 GPCRs comprise 20 receptor types that are activated by peptides, in addition to orphan receptors. Their ligand peptides include transmitters, adrenomedullin, calcitonin gene-related peptide, vasoactive intestinal peptide, and urocortins. Class-3 GPCRs include 11 metabotropic receptors that are activated by glutamate and γ-aminobutyric acid as well as orphan receptors. These receptors localize presynaptically to perivascular nerves, but are generally not expressed in endothelia and smooth muscles. Class-4 GPCRs incorporate receptors of the Frizzled and Smoothened families that are implicated in vascular remodeling. Many GPCR classes can be decomposed into GPCR subclasses and families (Tables 7.15 and 7.16). Each GPCR category contains a variable number of receptors. Synthesis of GPCR and their activity are tightly controlled. Availability of GPCRs relies not only on its production and exocytosis, but also on desensitization via endocytosis and resensitization by endosome recycling. Responsiveness of GPCRs depends on associated regulators, such as G proteins, kinases (feedback loops mediated by PKA and PKC) and phosphatases, as well as membrane partners. Ligand binding on G-protein-coupled receptors can be voltage sensitive, as GPCRs can serve as sensors for both transmembrane potential and external compounds [791] . Voltage-sensitive G-protein-coupled receptors are triggered by the voltage across the plasmalemma. 15 Gating currents act on structural GPCR components. G-protein-coupled receptors share a common structure of 7 membrane-spanning helices (transmembrane domains TM1-TM7) connected by 3 loops on each side of the membrane (intra-[IL1-IL3] and extracellular [EL1-EL3] loops, with an extracellular N-and intracellular C-terminus. Extra-and intracellular loops serve as binding sites for agonists (activators) and G proteins, respectively. The structure, conformation, and specificity of the G-protein binding site can depends on the ligand type. The ends of the third intracellular loop are involved in G-protein activation and selectivity of GPCR-G-protein interactions, as well as interactions with G-protein-coupled receptor kinases, arrestins, and other signaling molecules. Ligand binding causes a conformational change in the GPCR cytoplasmic domain. Class-A GPCR activation leads to an outward displacement of transmembrane helix 6 that opens for G-protein binding [792] . The activation rate of α2a-adrenergic 15. The resting potential difference across the 3-nm-thick plasmalemma is equal to about 70 mV. It can affect the conformation of membrane proteins. Table 7 .15. Classes of GPCRs. (Part 1) class A (GRH: gonadotropin-releasing hormone; TRH: thyrotropin-releasing hormone; Source: [790] ). Platelet-activating factor (PAF) receptor initiates cell response to its phospholipid ligand that has potent platelet aggregating and inflammatory effects and elicits smooth muscle contraction. Ligand family Amine Cathecolamines, dopamine, histamine, acetylcholine (muscarinic), serotonin G-protein-coupled receptors interact with guanine nucleotide-binding (G) proteins (Vol. 4 -Chap. 8. Guanosine Triphosphatases and Their Regulators) to transduce signals transmitted by messengers in the cardiovascular and ventilatory apparatus among others physiological systems. 16 Ligand-activated receptors catalyze the GDP-GTP exchange at a coupled G protein, and thereby promote the activity of effectors (second messenger-producing enzymes and ion channels). G-proteincoupled receptors thus act as guanine nucleotide-exchange factors (GEF) for G protein subunits. Release of GDP is the rate-limiting step in G-protein activation. After receptor activation, G protein binds to the receptor, the subsequent conformational change decreasing the affinity of the G protein for guanosine diphosphate. 16 . On the cardiomyocyte sarcolemma, angiotensin-2, endothelin-1, noradrenaline, and prostaglandin-F2α activate receptors coupled to a Gq subunit. Most receptors are able to activate more than one type of G protein. They thus trigger several signal transduction cascades. However, some receptors interact only with G-protein isoforms of the same class. Many GPCRs have a complex signaling behavior. In cardiomyocytes, β2-adrenoceptors couple to both stimulatory (Gα s ) and inhibitory (Gα i ) subunits that target adenylate cyclase as well as, upon phosphorylation by a G-protein-coupled receptor kinase, arrestins to signal via MAPK pathways . Mitogen-Activated Protein Kinase Modules) in a G-protein-independent manner. Desensitization of GPCRs involves multiple processes, such as receptor phosphorylation and arrestin-mediated endocytosis followed by receptor recycling or lysosomal degradation. Furthermore, GPCR oligomerization and localization to specific membrane compartments influence GPCR activity. G-protein-coupled receptors signal not only via heterotrimeric G proteins, but also various small monomeric GTPases to regulate the activity of effectors (Tables 7.17 to 7.19 ). Subunit Gα is composed of a GTPase and a helical domain [792] . The GTPase domain hydrolyzes GTP and provides the binding site for Gβγ dimer, GPCR, and effectors. It contains 3 flexible loops. The helical domain comprises 7 α-helices. Post-translational modifications of Gα regulate membrane location and between-protein interactions. All Gα, except Gα t , undergo palmitoylation at their N-termini. Certain Gα i are also myristoylated at their N-termini. Binding of GTP on a Gα subunit induces a structural rearrangement of the GPCR-G-protein complex that destabilizes this complex to interact with effector; some complexes dissociate, but not all. Subunits Gβ and Gγ form a functional unit. Several Gβγ dimers can interact with the same Gα isoform. Subunit Gβγ binds GPCRs and helps stabilize the GPCR-Gα interface. Subunit Gβ has a 7-bladed β-propeller structure with an αhelical N-terminus that forms a coiled-coil with N-terminus of Gγ [792] . The Cterminus of Gγ binds to blades 5 and 6. All Gγ undergo post-translational isoprenylation of their C-terminus with either a farnesyl (Gγ1, Gγ8, and Gγ11) or geranylgeranyl moiety. Subunits Gα and Gγ interact sequentially with activated GPCR to dock G protein onto the receptor. Activated Gα and Gβγ proteins positively or negatively regulate various effectors such as phospholipase-A, -C, and -D, 17 adenylate and guanylate cyclases, phosphoinositide 3-kinases, phosphodiesterases, protein kinase-C, and ion channels (Table 7 .20, Fig. 7 .1). 18 17. Phospholipases PLD1 and PLD2 regulate cytoskeletal organization and endo-and exocytosis. G-protein-coupled receptor signaling directs stimulation of PLD1 by PKC. 18. Transiently activated Gs-coupled β-adrenergic receptors cause a calcium influx through Ca V 1 channels that are phosphorylated by cAMP-dependent protein kinase-A [793] . (Mediator cAMP is produced in response to Gs activation.) On the other hand, sustained activation of β-adrenergic receptors causes their phosphorylation by GPCR kinases for recruitment of βarrestin-1 and endocytosis. Additional activation of Gi by β2-adrenergic receptors explains the difference in signaling by β1-(the predominant subtype in cardiomyocytes) and β2adrenergic receptors. Subunit Gi slows down the heart rate and atrioventricular conductance. Isoform Gi2, the major Gi subunit in cardiomyocytes, represses the activity of Ca V 1 channels. Table 7 .17. Subunit Gα of trimeric guanine nucleotide-binding proteins and their distribution and effectors (Part 1; Source: [767] ; ACase: adenylate cyclase; ACi: adenylate cyclase isoform i; Ca V : voltage-gated Ca ++ channel; GIRK: Gβγ-regulated inwardly rectifying K + channel [K IR 3]; G Tc , G Tr : cone and rod transducin; PDE: phosphodiesterase). Gα subunits define the basic properties of a heterotrimeric G protein. They can be classified into 4 families, Gα s , Gα i/o , Gα q/11 , and Gα 12/13 . In addition to Gs, 2 transcripts that encode XL αs and neuroendocrine secretory protein NESP55 are generated by promoters upstream of the GNAS (Gs-encoding) promoter of the GNAS complex locus. Extra-large protein Gs XL that has a limited expression pattern (adrenal gland, heart, pancreas, brain, and pituitary pars intermedia) is able to bind to Gβγ dimer and provoke cAMP synthesis. The GNASXL gene also produce ALEX that inhibits Gs XL . In addition, exon 3 of the GNAS gene is alternatively spliced to give rise to long (Gs L ) and short (Gs S ) Gs isoforms. Subsequently, these effectors activate or inhibit the production of second messengers (cAMP, cGMP, diacylglycerol, inositol trisphosphate, phosphatidylinositol Go-Coupled muscarinic channels M 2 also inhibit Ca V 1 channels. In enteroendocrine cells, bitter taste receptors (T 2 R) and Gα gust of the Gi/o group increase Ca ++ influx via activated Ca V 1 channels [794] . In the hippocampus, activated Gi/o-coupled adenosine receptor A 1 suppresses serotonin release, as it inhibits Ca V 2.2 and Ca V 2.1 channels (more precisely Ca V 2.2-PKC-syntaxin and Ca V 2.1-PKA-synaptobrevin axes) [795] . On the other hand, activated Gscoupled A 2 stimulates serotonin release, as it stimulates Ca V 2.1 channels (Ca V 2.1-PKAsynaptobrevin axis). The 3 Gi subtypes (Gi1-Gi3) activate K + channels K IR 3 (K ACh current) of atriomyocytes (Gβγ dimer is inactive) [796] . Gi/o-Coupled glutamate receptor mGluR7 is able to inhibit Ca V 2.1 channels via Gβγ dimer. Upon Ca ++ influx, Ca ++ -calmodulin binds to macrophage myristoylated alanine-rich C-kinase substrate (macMARCKS) that is tethered to mGluR7 [797] . It then displaces macMARCKS and Gβγ from mGluR7 receptor. Gβγ Dimer can then inhibit Ca V 2.1 channel. In sympathetic neurons, Gβγ inhibits Ca V 2.2 channels upon stimulation by noradrenaline and somatostatin [798]. ). Subunits G15 and G16 are the murine and human ortholog, respectively. Enzymes PLCβ1, -3, and -4 are the major effectors of Gq/11 and PLCβ2 is a minor effector. Subunits G12 and G13 are often activated by Gq/G11-coupled receptors (crosstalk). Cadherins (a portmanteau word for calcium-dependent adhesion) are type-1 transmembrane proteins and constituents of adherens junction. They are connected to nearby actin filaments via α-actinin and cortical actin via catenin and vinculin. Radixin is a cytoskeletal linker of actin to the plasma membrane. Members of the Gα i , Gα q , and Gα 12/13 families cooperate to regulate Ca ++ signal and nitric oxide (NO). Subunit Gα q targets phospholipase-C and causes Ca ++ influx without the assistance of NO. Subunit Gα 13 supports Ca ++ influx by a mechanism dependent on NO, guanylate cyclase, and cGMP (a process blocked by activated adenylate cyclase, the cAMP-PKA axis preventing Ca ++ influx and NO production). NO may relieve inhibition of adenylate cyclase caused by Gi subunit via adpribosylation of Gi subunit. Distribution Effectors (3,4,5)-trisphosphate, and arachidonic and phosphatidic acids), and promote calcium influx, and opening or closure of various ion channels. A relatively small number of G proteins transduce signals from a large number of GPCRs. Each member of the G-protein family is thus able to interact with many different GPCRs. In addition, many GPCRs can activate multiple G proteins. Suitable signal transduction relies on specific interactions. Each contact site of Gα subunits, as well as other molecular regions, contribute to coupling specificity [792] . In addition, specific isoforms of Gβ interact preferentially with specific GPCRs. 19 The 19. Subunits Gβ4 and Gβ5 have the strongest and the poorest coupling ability with β1adrenergic and A 2A adenosine receptors. Subunits Gβ1 and Gβ4 have a similar coupling capacity with β1-adrenergic receptor, but Gβ1 has 20-fold lower coupling capacity than Gβ4 for adenosine receptor. [767] ). In the absence of signaling, Gα subunit is tethered to its associated Gβγ dimer. This dimer is assembled from a repertoire of 5 Gβ (Gβ1-Gβ5) and 12 Gγ (Gγ1-Gγ5, Gγ7-Gγ8, and Gγ10-Gγ14) subunits. Subunit Gβ5 only binds weakly to Gγ and links to regulators of G-protein signaling. It resides mainly in the brain. Subunits Gβ1 to Gβ4 as well as Gγ2, Gγ4 to Gγ6, and Gγ10 to Gγ12 are widely distributed. Subunits Gγ1, Gγ3, and Gγ13 lodge in the central nervous system. Subunit Gγ8 resides in the olfactory epithelium and Gγ3 also in blood cells. The Gα and Gβγ subunits stimulate various effectors, such as adenylate and guanylate cyclases, phosphodiesterases, phospholipase-A2 and -C (PLC), phosphoinositide 3-kinases (PI3K), besides guanine nucleotide-exchange factor (GEF), thereby activating or inhibiting the production of multiple second messengers such as cAMP, cGMP, diacylglycerol (DAG), inositol (1,4,5)-trisphosphate (IP 3 ), Ca ++ ions, phosphatidylinositol (3,4,5)-trisphosphate (PIP 3 ), arachidonic and phosphatidic acids, and opening or closing of numerous ion channels. Mediators PLC and PI3K via different pathways activate the MAPK module. Adenylate cyclases AC1 (−), AC2/4/7 (+) Ion channels Phospholipases PLCβ1-PLCβ3 (+) (in increasing order of potency) (DAG-PKC-cRaf-MAPK, IP 3 -Ca ++ -RasGRF-Ras-cRaf-MAPK, cAMP-PKA-RapGEF3/4-Rap-bRaf-MAPK axes) subtype of Gγ influences the linkage of the Gβγ dimer. 20 Furthermore, homologous Gα binding to GPCRs depends on the GPCR ligand. 21 Proteic constituents of ubiquitous primary cilia are required for the localization of G-protein-coupled receptors, at least on central neurons [799] . Certain GPCRs such as somatostatin receptor SstR3 and serotonin receptor-6 are specifically located in neuronal cilia. Ciliary localization of somatostatin receptor SstR3 (neurotransmission) and melanin-concentrating hormone receptor MchR1 (feeding regulation) in neurons needs ciliary disorder Bardet-Biedl syndrome BBS2 and BBS4 proteins. G-protein-coupled receptors also act as scaffolds for the formation and location of signaling complexes in the cell. The response properties are determined by the relationship among the ligand, receptor, G protein, and associated proteins. Recep-20. Dimers Gβ1γ5 and Gβ1γ7 can mediate binding to M 2 muscarinic receptor, but not Gβ1γ2. 21. Activated peptidase-activated receptor-1 functions according to ligand type. For example, thrombin favors Gα q rather than Gα 12/13 . tor signaling is regulated by both endogenous and exogenous actions. Activation of G-protein-coupled receptors can be sensitive to the plasmalemma lipid composition. Local cholesterol and sphingolipid concentrations affect the ligand binding and receptor transport. Although ubiquitous GPCRs classically signal via heterotrimeric G proteins, they can also signal via β-arrestins, even without the participation of G proteins. β-Arrestin adaptors can control receptor signaling, desensitization, and trafficking. Most GPCRs use both β-arrestins and G proteins. However, some GPCR receptors that fail to activate G protein can act as coreceptors or decoy receptors that eliminate ligands. Agonist binding can prime β-arrestin association with decoy receptor CXCR7, hence β-arrestin-dependent signaling via activation of mitogen-activated protein kinase in the absence of G-protein. Therefore, GPCRs can signal exclu-sively via β-arrestins (non-canonical pathway). Ligand CXCR7 stromal-derived factor SDF1α initiates β-arrestin-2-mediated endocytosis, whereas another ligand, interferon-inducible T-cell α chemoattractant fails to activate Gi and Gq, but promotes vascular smooth muscle cell migration [800]. Many G-protein-coupled receptors exhibit a basal activity in the absence of their agonists with a variable magnitude according to GPCR type. G proteins can interact with GPCRs before ligand binding (GPCR-G-protein precoupling). The ligandindependent constitutive activity allows the conformation change to an active state upon stimulation, owing to interactions with the plasmalemmal lipids. The plasmalemma tension can then modulate the GPCR activity. The GPCR stimulation then catalyzes G-protein activation. The ligand-receptor complex is supposed to constitute the functional unit of GPCR signaling. However, G-protein-coupled receptors can form homo-or heterodimers. Moreover, some GPCRs form higher-order oligomers upon homo-or heterodimerization during their exocytosis, activation, inactivation, and/or endocytosis. Transient or sustained di-and oligomerization may explain the negative or positive receptor cooperativity. Therefore, G-protein-coupled receptors operate as monomers as well as unstable dimers or oligomers. Agonist of a GPCR then must choose one protomer of the GPCR complex, its binding influencing the activity of associated protomers, among which at least one functional unit activates signaling effectors. The history of GPCR protomer association and dissociation from synthesis to degradation on cell signaling may play a role. Heterodimerization can lead to signaling complexes that have a single ligand binding. However, only one GPCR of the dimer can be active in signaling. Two subunits of a receptor dimer coupled to G protein differ in their conformation [801] . A receptor dimer with a single ligand-bound subunit can trigger G-protein activation. The interaction of G protein with the receptor dimer prevents a symmetrical functioning of the dimer and explains the negative cooperativity in ligand binding on GPCR dimers. Both class-A and class-C GPCRs (Sect. 7.5) can act as oligomers [802] . Metabotropic glutamate receptors assemble into strict dimers. γ-Aminobutyric acid receptor-B spontaneously forms dimers of heterodimers. Homo-and heterodimerization of GPCRs exist for class-C receptors, not only metabotropic γ-aminobutyric acid (GABA B ) and glutamate (mGluR) receptors [803], 22 but also taste heterodimeric-(T 1 R1-T 1 R3) and calcium-sensing receptors, for which only dimers are involved in signal transduction. Whereas, at least, some class-A GPCRs may function as monomers, other class-A GPCRs for glycoproteic hormones, such as luteinizing, follicle-stimulating, and thyroid-stimulating hormone, may transduce their signal in vitro as di-and/or oligomers by transactivation. In vivo, intermolecular cooperation can rescue GPCR defective in binding as well as signaling capacity [804] . Intermolecular cooperation and di-or oligomerization between mutant binding-and signaling-deficient luteinizing hormone receptors, indeed, re-establish luteinizing hormone effect in the absence of functional wild-type receptors. In the cardiovascular system, G-protein-coupled receptors regulate numerous functions, such as heart frequency and contractility as well as vascular tone. More than 100 different GPCRs are involved in the cardiovascular system, such as (muscarinic) acetylcholine, adenosine, adrenergic, angiotensin-2, bradykinin, endothelin, lysophosphatidic acid, serotonin, sphingosine 1-phosphate, and vasopressin receptors. G-protein-coupled receptors abound in the vasculature, particularly those of class-1 (Vol. 5 -Chap. 7. Vessel Wall). In blood vessels, class-1 GPCRs, once they 22. Metabotropic glutamate receptors are constitutive dimers that do not form larger oligomers. Like receptor Tyr kinases and receptor guanylate cyclases, agonist stimulation of mGluR dimer with a protomer that has its extracellular agonist-binding domain uncoupled from its G-protein-activating transmembrane region causes a symmetric activation of twin transmembrane domains with the same efficiency [803] . Agonist binding to one Venus flytrap (VFT) extracellular domain not only cis-activates the transmembrane domain of the same subunit, but also trans-activates that of the other protomer owing to an intersubunit rearrangement. are activated by chemical messengers that comprise peptides act directly as vasoconstrictors or vasodilators or indirectly as vasodilators. 23 Smooth muscle tone is controlled by the phosphorylation state of the regulatory myosin-2 light chain (MLC). Phosphorylation of MLC, hence the vasomotor tone, depends on intracellular calcium concentration. Vasodilation results from: (1) low intracellular calcium concentration by Ca ++ export (egress from cytosol by reuptake into the sarcoplasmic reticulum and expulsion across the plasma membrane) through Ca ++ carriers and (2) MLC dephosphorylation by myosin light chain phosphatase (MLCP) under the control of RoCK kinase (Table 7.21) . Conversely, vasoconstriction results from increased Ca ++ concentration that, in cooperation with calmodulin, activates Ca ++ -calmodulin-dependent myosin light chain kinase (MLCK). The latter stimulates cross-bridge cycling of actin-myosin stress fibers. Vasodilation can be primed by: (1) trans-plasmalemmal hyperpolarization (e.g., on elevated activity of ATP-sensitive K + channel) that precludes the activity of voltage-gated calcium channels (mechanism used by adenosine) and (2) cAMP and cGMP signaling that stimulates protein kinase-A and -G, respectively, that both phosphorylate MLCK (procedure utilized by prostaglandins [PGD2, PGE2, and PGI2] and nitric oxide, respectively). PKA Kinase also hampers the activity of RhoA GTPase. Vasoconstriction can be intiated by: (1) trans-plasmalemmal depolarization that promotes the activity of voltage-gated calcium channels (this process can be further enhanced by voltage-gated Na + channels and is typically triggered by ATP and tension); (2) decline in PKA activity by Gi subunit; and (3) elevated PLC activity by Gq subunit that causes, via IP 3 , Ca ++ influx, hence MLCK activation, as well as RhoA stimulation (thus RoCK excitation that hinders MLCP action). 23. Blood pressure-lowering drugs target a limited number of class-1 GPCRs. Such drugs include angiotensin-2 receptor and adrenoceptor antagonists. Vasoconstrictors include adrenaline, angiotensin-2, apelin, adenosine triphosphate, endothelin-1, motilin, neuromedin-U, neuropeptide-Y, sphingosine 1-phosphate, thrombin, thromboxane-A2, urotensin-2, and vasopressin [805] . Vasodilators encompass extracellular adenosine, extracellular ADP and ATP, bradykinin, ghrelin, histamine, natriuretic peptides, nitric oxide, noradrenaline (via β2-adrenergic receptors), platelet-activating factor, some prostaglandins, as well as substance-P ghrelin. Ghrelin targets growth hormone secretagog receptor and nociceptin. On vascular smooth muscle cells, GPCRs are predominantly coupled to Gs. They thus activate adenylate cyclase and cause direct vasodilation. Among class-2 GPCRs that are activated by peptides, all those with vasoactivity directly operate as vasodilators. They are targeted by adrenomedullin and urocortins. 24 On vascular endothelial cells, GPCRs can generate and release: (1) nitric oxide; (2) arachidonic metabolites such as prostacyclin; or (3) endothelium-derived hyperpolarizing factor that target adjoining smooth muscle cells, where they excite soluble guanylate cyclase, Gs-coupled prostanoid IP 1 receptors, and induce hyperpolarization, respectively. All these processes create vasodilation. Cardiac regulation by the sympathetic nervous system is carried out by βadrenergic receptors that are coupled primarily to Gs. Produced cAMP targets hyperpolarization-activated, cyclic nucleotide-gated channels and activates protein kinase-A. The latter phosphorylates Ca V channels, phospholamban, and troponin-I [767] (Vol. 5 -Chap. 5. Cardiomyocytes). These events explain positive chronotropic (C+; increased heart rate), dromotropic (D+; elevated conduction velocity through the nodal tissue), lusitropic (L+, enhanced cardiomyocyte relaxation), and inotropic (I+; improved contractility) effects of the sympathetic command. On the other hand, the parasympathetic nervous system has negative chronotropic (C−) and dromotropic (D−) effects. It operates via muscarinic acetylcholine M 2 Gi/o-coupled receptor. In addition to the inhibition of cAMP production, this receptor activates atrial G-protein-regulated inward rectifier K + channels (GIRK; K IR 3.1 and K IR 3.4) via released Gβγ dimer [767] . Muscarinic receptors also inhibit voltagegated Ca V channels. Like in vascular smooth muscle cells, the tone of airway smooth muscle cells is mainly regulated by members of the Gq/11 and Gs families of G-protein subunits that cause bronchoconstriction and bronchodilation, respectively. 24. G-protein-coupled receptors on vascular smooth muscle cells that provoke vasoconstriction can be linked to more than one signaling cascade via different families of G-protein subunits (Gi/o, Gq/11, and/or G12/13) . Subunits of the Gi/o, Gq/11, and/or G12/13 families activate primarily, but not exclusively, phospholipase-C and small monomeric GTPase Rho and inhibit adenylate cyclase, respectively [805]. Acetylcholine released from postganglionic parasympathetic nerves intervenes mainly via Gq/11-coupled M 3 receptors. Gq/11-coupled receptors in airway smooth muscles also comprise H 1 histamine, B 2 bradykinin, ET B endothelin, and CysLT 1 leukotriene receptor [767] . Subunit Gi impedes the relaxation of airway smooth muscle cells. Gs-coupled receptors provoke relaxation of contracted airway smooth muscle cells, such as β2-adrenoceptor, EP 2 prostaglandin, and IP prostacyclin receptor [767]. Platelet adhesion and activation is initiated by interaction with components of the subendothelial matrix, such as collagen and von Willebrand factor (Vol. 5 -Chap. 9. Endothelium). Collagen is able to cause a firm adhesion of thrombocytes to the subendothelial layer. Additional platelets are recruited by diffusible mediators, such as ADP, ATP, and thromboxane-A2, that are secreted by activated thrombocytes. G-Protein-coupled receptors operate in platelet aggregation. Twelve most abundant platelet GPCRs include: (1) thrombin receptor, i.e., peptidase-activated receptors PAR 1 and PAR 4 ; 25 (2) adenosine receptors A 1 , A 2A , and A 2B ; (3) nucleotide receptors P2Y 1 , P2Y 12 , as well as P2Y 10 ; (4) GPR183 26 (5) succinate receptor-1 (SucnR1 or GPR91); (6) lysophosphatidic acid receptors LPA 1 , LPA 3 , LPA 4 (GPR23), and LPA 5 (GPR92); (7) α2a-adrenergic receptor; (8) glutamate mGlu 3 and mGlu 4 ; and (9) serotonin receptors 5HT 1F and 5HT 4 [836] . Agent ADP activates the G-protein subunits Gq and Gi via P2Y 1 and P2Y 12 receptors, respectively. Thromboxane-A2 targets its cognate Gq-coupled receptor TP. Thrombin binds to peptidase-activated receptors that are coupled to Gq, G12/G13, and in some cases to Gi [767] . Adrenaline connects to Gz-coupled α2-adrenergic receptors to potentiate the effect of other platelet activators. Leukocytes move according to a concentration gradient of chemoattractants. They can then enter into lymphoid organs to mature and, then, exit for antigen surveillance. They can also reach sites of infection. Lymphocyte chemoattractants include chemokines, sphingosine 1-phosphate, and lysophosphatidic acid. Chemokine receptors act mainly via Gi subunits. Lysophospholipid receptors activate Gi, G12/G13, and Gq/G11 subunits. Hematopoietic cells are able to synthesize Gq, G11, and G16 subunits. Members of the G12/13 family target RhoA GTPase. Neutrophils respond to diverse chemoattractants, such as N-formyl Met-Leu-Phe (fMLP), C5a, platelet-activating factor, or interleukin-8. Complement C5a element 25. Activated receptor PAR 1 is more potent than PAR 4 . 26. A.k.a. Epstein-Barr virus-induced gene product-2 EBI 2 . Epstein-Barr virus-induced gene product-1 (EBI 1 ) is the (CCL19 and CCL21) chemokine receptor CCR7. Up-and downregulation of GPCRs and their ligands result from viral infections. operates via Gi2, Gi3, and Gβ2 subunits to stimulate PI3Kγ as well as PLCβ2 and PLCβ3. In addition, PIP 3 -dependent Rac exchanger-1 (PREx1) that acts as a guanine nucleotide-exchange factor for small GTPases Rac, is boosted by Gβγ dimer. Mastocytes also express various GPCRs that intervene during allergy and anaphylaxis (with vasodilation, bronchoconstriction, etc.) . Ligands, such as anaphylatoxins C3a and C5a, leukotrienes, sphingosine 1-phosphate, and platelet-activating factor mediate anaphylaxis, acting as transmitters within mastocytes and extracellular medium, including circulating biofluids. Pulmonary mastocytes express a huge number of GPCRs, such as [837]: (1) complement component receptors C3aR1 and C5aR1; (2) cysteinyl-leukotriene receptor CysLT 1 ; (3) sphingosine 1-phosphate receptor S1P 1 ; (4) corticotropin-releasing hormone (CRH) receptor CRF 1 ; (5) plateletactivating factor receptor (PAFR); (6) cadherin, EGF LAG seven-pass receptor CELSR1; 27 (7) Zn ++ -activated GPR39; (8) progestin and adiponectin, C1q, and collagen domain (adipoQ)-containing receptor family member PAQR5; (9) lysophosphatidic acid receptor LPA 5 ; (10) succinate receptor-1 (SucnR1); and (11) GPR12. Acupuncture is an old medical technique in traditional chinese medicine. In acupuncture, the internal organs are assumed to be interconnected by meridians, i.e., pathways in which the vital energy (Qi) flows throughout the body. Thin needles are manipulated (using lifting, thrusting, and rotation) to stimulate specific points (acupoints) of the body to restore the balance between Yin and Yang energy by removing blocks in the flow of Qi [806] . 28 The effects of acupuncture can be explained by interactions between the nervous, circulatory, endocrine, and immune systems. The subcutaneous connective tissue is composed of cells embedded in the extracellular matrix mainly constituted by collagen and elastic fibers in a gel of glycoproteins and proteoglycans. Mastocytes can be presumed to play a major role in acupuncture. They are scattered, but localize predominantly near blood and lymphatic vessels and nerves. Their density is higher near acupoints [809] . A mechanical stress field results from local deformations of the connective tissue imposed by the needle motions. This stress field is sensed by the local population of mastocytes that react, in particular, by degranulation. Released molecules include calcitonin gene-related peptide (CGRP), heparin, histamine, leukotrienes (LTb4, LTc4, LTd4, and LTe4), platelet-activating factor, prostaglandin-E2, serotonin, substance-P, and thromboxane-A2 (Table 7 .22). Mastocytes also secrete pep-27. A.k.a. Flamingo homolog-2. 28. The acupuncture needle is inserted in the subcutaneous connective tissue and manipulated until a special sensation is obtained, which is supposed to prove that the needle has been placed in the proper location. The same acupoints can also be stimulated by other methods, such as moxibuxion, acupressure, and electroacupuncture. Moxibustion relies on heating acupoints by burning a moxa stick made of Artemisia vulgaris (common mugwort) to stimulate the blood circulation and produce a smoother flow of Qi [807] . Acupressure can be referred as needleless acupuncture by applying finger pressure. Electroacupuncture relies on the electrical stimulation of acupoints [808]. Table 7 .22. Released molecules by the mastocyte and their effects. Acupuncture can be modeled by an immediate and a late response. Nerves and mastocytes exchange chemical messengers such as substance-P. The latter stimulates histamine and nitric oxide (NO) release. Calcitonin gene-related peptide (CGRP) causes a vasodilation; nitric oxide cooperates with CGRP to increase its positive inotropic effect that raises the local blood flow in dilated vessels. Histamine is quickly catabolized, thereby acting near the site of release. Resulting vasodilation and increased vessel wall permeability support the transfer of chemical mediators into the blood circulation. The NO concentration rises and enhances the vasodilation. Serotonin has a biphasic effect, as it triggers a vasoconstriction and promotes NO release, hence a subsequent vasodilation. Nerve growth factor (NGF), tumor-necrosis factor (TNF) and interleukins (IL) are potent mastocyte chemoattractants. Mastocyte chemotaxis is supported by matrix degradation by secreted peptidases. Vasoconstriction followed by NO-mediated vasodilation Thromboxane-A2 Vasoconstriction, platelet aggregation Tryptase, chymase Matrix degradation for enhanced cell migration tidases (e.g., tryptase), growth factors (e.g., FGF, gmCSF, and NGF), and cytokines (e.g., interleukins and tumor-necrosis factor). Nerve endings are stimulated and release substance-P that further activates mastocytes and triggers the production of nitric oxide. Meridian can be assumed to be a neurovascular signaling tract. A compartmental model can then be designed. Compartment 1 is the acupoint region with its 3 components: (1) mastocytes, (2) blood and lymph vessels, and (3) nerves. Chemoattractants augment the mastocyte population (auto-amplification); autocrine signals bestow a self-sustained response. Nerves and mastocytes exchange cues. Compartment 2 is related to signal transmission to the central nervous system, either very rapidly via nervous impulses, or delayed via messenger convection through the blood circulation. A feedforward loop associated with elevated cardiac function enables an increased blood flow for a relevant material transport. Compartment 3 deals with signal processing with a quick and late responses corresponding to fast and delayed inputs. Compartment 4 represents outputs sent from the central nervous system and the body's response. G-protein-coupled receptors not only act via heterotrimeric G proteins, but can also function in a G-protein-independent manner. β2-Adrenergic receptors signal via either Gα or via tyrosine kinase Src according to the ligand concentration, low or high, respectively [810] . Activated Gi-coupled receptors often contribute to inositol phosphate signal triggered by Gq-coupled receptors (synergistic receptor crosstalk). For example, Gicoupled adenosine A 1 and α2c-adrenergic receptors collaborate with Gq-coupled bradykinin B 2 and P2Y receptors. Gβ1γ2 Dimer as well as combinations of Gβ (Gβ1-Gβ3) with Gγ (Gγ2-Gγ7) increases potency of bradykinin or UTP messenger without interfering with Gα q signaling [811] . This GPCR crosstalk results from Gβγ exchange between Gi-and Gq-coupled receptors. Certain GPCR effectors do not depend on G proteins. Crosstalk between GPCRs and small GTPases exists. 29 GPCR signaling through G proteins can activate Ras and Rho GTPases. Activated Ras triggers the mitogen-activated protein kinase cascade. RhoA, Rab, Arf, and ArfGEF can directly associate with GPCRs. GPCRs can also function as guanine nucleotide-exchange factors for small GTPases. The interactions between GPCRs and small GTPases is required for cellular transport and cell migration. GPCRs are synthesized and modified in the endoplasmic reticulum, then transported to the Golgi body for additional changes, lastly to the plasmalemma. Rab GTPases are implicated in exocytosis. Rab GTPases also regulate GPCR endocytosis into early endosomes, GPCR targeting to lysosomes for degradation, or GPCR recycling from early endosomes. Arf1 and Arf6, with Arf GEFs (adpribosylation factor nucleotide-binding site opener [ARNO] ) and β-arrestin, also control GPCR endocytosis [812] . Activated Gi-coupled chemoattractant receptor, the N-formylmethionyl-leucylphenylalanine receptor (fMLPR), leads to both Ras-dependent and -independent activation of Ral GTPase. Ral GEF RalGDS interacts with Ras, which activates Ral, hence, actin cytoskeleton reorganization (Ras-dependent mechanism). Plasmalemmal translocation of complexes made by β-arrestin and RalGDS allows binding to fMLPR and uncoupling from G protein, thus activates membrane-bound Ral (Rasindependent mechanism) [812] . Ligand binding to GPCRs activates small GTPases either directly or indirectly. 30 Rho-dependent responses can be activated by Gq and G12 ( Fig. 7.2) . Furthermore, G12 can bind and activate Rho-specific guanine nucleotide-exchange factors [813] . Mutant Gs-induced cAMP can activate small GTPases Rap1 and B-Raf that, in 29. The small GTPase superfamily includes at least five families: Ras GTPase subfamily (Ras, Rap, and Ral) of regulators of cell signaling, Rho GTPase subfamily (Rho, Rac, and Cdc42) of regulators of actin cytoskeleton, Rab GTPase subfamily of regulators of vesicular transport, Arf GTPase subfamily (Arf, Arl, and Sar) of regulators of vesicular transport, and Ran GTPase subfamily of regulators of microtubule organization and nucleocytoplasmic transport. 30. GPCRs for lysophosphatidic acid and thrombin induce stress fibers, focal adhesions, and cell rounding through Rho-dependent pathways [813] . (2) Ras via PLCβ and activation of (2a) Ca ++dependent Ras guanine nucleotide-releasing factor (RasGRF) and (2b) Ca ++ -and PKCregulated proline-rich tyrosine kinase-2 (PYK2) of the focal adhesion kinase family. GPCR stimulation also leads to phosphorylation of Src homology and collagen (SHC), which subsequently forms complexes with growth factor receptor-bound-2 (GRB2), and activation of tyrosine kinases acting on Son of sevenless (Sos Stimulation of GPCRs can stimulate receptor Tyr kinases, such as epidermal growth factor receptors, via: (1) proteolytic cleavage by ADAM metallopeptidase and release of EGF-like ligands, such as heparin-binding EGF-like growth factor, and (2) NRTK activation that can phosphorylate EGFR tyrosine residues. Activation by GPCRs of the Ras-MAPK pathway uses alternate RTK-dependent and -independent pathways according to cell types. Angiotensin-2 in thoracic aortic smooth muscle cells requires RTK activity to signal to ERK, but not in smooth muscle cells of the renal microvasculature [815] . β2-Adrenergic receptor mediates extracellular signal-regulated kinase activation via assembly of a multireceptor complex with the epidermal growth factor receptor following co-internalization of both receptors into clathrin-coated vesicles [816] . Several NRTKs (CSK, Lyn, BTK, proline-rich tyrosine kinase-2, and focal adhesion kinase; Vol. 4 -Chap. 3. Cytosolic Protein Tyr Kinases) could mediate MAPK activation by Gi and Gq [768] . Src or Src-like kinases can phosphorylate SHC on stimulation by Gβγ and α-adrenoceptors, after recruitment of β-arrestin and GRK2 kinase. Kinase Src can be activated also by interaction with Gi and Gs or β3-adrenoceptors. Subunit Gq can stimulate components of the MAPK module via: (1) a PKCdependent, Ras-independent mechanism; (2) PKC-and Ras-dependent process; or (3) PKC-independent, Ras-dependent procedure, depending on the cell type and receptor expression level. Isoforms of PI3K kinase can be required for GPCR-induced MAPK activation ( Fig. 7 .2). Subtype PI3Kγ is activated by direct interaction with Gβγ subunit. It can then act on the SHC-GRB2-SOS-Ras pathway. The Gβγ dimer activates PI3Kβ, which then stimulates PKB [817] . Arrestins bind many phosphorylated GPCRs for endocytosis. β2-Adrenoceptors can associate with the Na + -H + exchanger regulatory factor (NHERF); angiotensin receptors AT 1A with Janus kinase JaK2; metabotropic glutamate receptors with Homers, which can complex with IP 3 receptors; muscarinic acetylcholine M 3 receptors and AT 1A with GTPases Rho and ARF [768] . GPCR ligands, such as lysophosphatidic acid, endothelin-1, platelet-activating factor, and thrombin can induce a rapid transient phosphorylation of epidermal growth factor receptors in fibroblasts and in vascular smooth muscle cells. Catecholamines, angiotensin-2, and endothelin-1 act in cardiomyocytes where they can activate heparin-binding epidermal growth factor with members of a disintegrin and metallopeptidase (ADAM) family and cause GPCR-induced cardiac hypertrophy ( Fig. 7.3) . Different types of G proteins (Gi, Gq, and G12), different metallopeptidases of the ADAM family (ADAM10, ADAM12, and ADAM17), and different EGF-like ligands (heparin-binding epidermal growth factor, transforming growth factor-α, and amphiregulin) are involved in inter-receptor crosstalk (or transactivation) [818]. Regulators of G-protein signaling are GTPase-accelerating proteins that activate the GTPase activity of α subunits of heterotrimeric G proteins, thereby inactivating G protein and rapidly switching off the GPCR signaling pathway. All the RGS proteins contain an RGS domain. Small RGS proteins, such as RGS1 and RGS4, are slightly greater than the RGS domain. Other RGSs contain additional Crosstalk between receptors of angiotensin-2 (AT 1 ) and endothelin-1 (ET A , or ETRA) and epidermal growth factor receptor (EGFR) in the cardiomyocyte. Transactivation of EGFR requires metallopeptidases of the ADAM family, particularly ADAM12, heparinbinding EGF-like growth factor, docking molecule GRB2-binding protein GAB1, Src Tyr kinase, and phosphoinositide 3-kinase. Transactivation of EGFR leads to MAPK activation. domains that confer further functionality, such as DEP for membrane targeting, GGL (G-protein-γ subunit-like) for binding Gβ subunits, GoLoco for guanine nucleotide inhibitory activity, PH for guanine nucleotide-exchange stimulating effect, and PDZ for GPCR, PTB for phosphotyrosine, PX for phosphatidylinositol, and RBD for Ras binding. Regulators of G-protein signaling can also speed up the onset of signaling, in addition to accelerating deactivation without changing amplitude or sensitivity of the signal. This paradoxical activity has been attributed to enzymatic (GAP) or scaffolding (non-GAP) functions. Yet, fast GTPase activity of RGSs may suffice to explain the activation kinetics and agonist sensitivity, i.e., increased onset, deactivation rates, and blunting of sensitivity [819]. Agonist-GPCR binding strongly enhances interactions between GPCRs and G proteins, G-protein-coupled receptor kinases (GRK), and β-arrestins. Agonists of GPCRs can simultaneously activate G-protein-and β-arrestin-dependent pathways. Agonist-activated GPCRs are also phosphorylated by G-protein-coupled receptor kinases. Phosphorylation of GPCRs by GRKs decreases GPCR interactions with G proteins and increases GPCR binding of β-arrestins (Sect. 7.11). β-Arrestins can then further desensitize GPCR-mediated signaling by several mechanisms: (1) receptor hindrance due to β-arrestin attachment; (2) recruitment of second messenger-degrading enzymes; and (3) scaffold proteins that facilitate receptor endocytosis. However, β-arrestin binding can initiate additional signaling programs via recruitment of signaling mediators such as mitogen-activated protein kinases to activated receptors. Therefore, interaction of GPCRs with β-arrestins prevents GPCR signaling via G proteins, but can simultaneously prime other signaling pathways as well as associate active receptors to clathrin-coated pits to facilitate receptor endocytosis that controls GPCR desensitization and resensitization, in addition to their endocytosismediated activity. Ubiquitous G-protein-coupled receptor kinases constitute a 7-member family (GRK1-GRK7). These kinases that have different distribution patterns among the body's tissues as well as distinct binding preferences for some receptors. They can yield functional specialization. Isoforms of GPCR kinases determine β-arrestin function, as they can lead to functionally distinct pools of β-arrestin, possibly by phosphorylating distinct receptor sites. Enzymes GRK5 and GRK6 are required for βarrestin-mediated MAPK signaling. G-protein-coupled receptor kinases, particularly GRK2 isoform, not only target G-protein-coupled receptors, but also numerous other substrates, such as receptor Tyr kinases (PDGFR and EGFR), as well as members of the GIT family of GTPaseactivating proteins of adpribosylation factor (ArfGAP), DM2 ubiquitin ligase, and catalytic subunit P110 of PI3K, PKB, MAP2K, and P38MAPK kinases. They also have many interacting proteins. The GRK2 subtype can attenuate PDGF-dependent proliferation of smooth muscle cells, but favors mitogenic signaling induced by EGF in osteoblasts and the Hedgehog receptor Smoothened in fibroblasts [820] . In addition, increased GRK2 concentration potentiates migration of epithelial cells directed by fibronectin and sphingosine 1-phosphate. G-protein-coupled receptor kinases possess their own regulators. Calcium-binding recoverin is a neuronal calcium sensor that localizes to the retina and serves as a Ca ++ -dependent regulator of GRK1 kinase. At high Ca ++ concentration, recoverin sequesters GRK1, whereas at low Ca ++ level, the complex dissociates and GRK1 becomes active. In response to GPCR stimulation, GRK2 is rapidly phosphorylated by Src (Tyr13, Tyr86, and Tyr92) and MAPK (Ser670) kinases and then degraded by the proteasome [820] . Cell cycle progression requires changes in the activity or concentration of signaling proteins. The GRK2 kinase is phosphorylated (Ser670) by cyclin-dependent kinase CDK2 during the G2-M transition and subsequently binds to peptidyl-prolyl isomerase Pin1 to be degraded [820]. G-protein-coupled receptors experience cycles of phosphorylation and dephosphorylation. Membrane-associated G-protein-coupled receptor phosphatase (GRP) aims at dephosphorylating GPCRs that have been phosphorylated by G-protein-coupled receptor kinases to cause agonist-dependent receptor deactivation, hence restoring the ground state of the receptor. 31 A family of receptor phosphatases contribute to the regulation of G-proteincoupled receptors. G-protein-coupled receptor phosphatase is an unsual form of protein phosphatase-2 with peculiar subcellular distribution and substrate specificity [822] . Desensitization of GPCRs is initiated by phosphorylation of Ser and Thr residues of intracellular loops and C-terminus by second messenger-activated kinases, such as protein kinase-A and GPCR kinases. Once phosphorylated, GPCRs generally bind to arrestin that prevents subsequent coupling between GPCR and G-protein and elicits GPCR integration into clathrin-coated pits for endocytosis. Moreover, when arrestins bind to GPCRs with high affinity, they preclude phosphatase action on receptors. On the other hand, endocytosis can be a prerequisite for resensitization of Gprotein-coupled receptors, such as for β2-adrenoceptors, when dephosphorylation occurs in endosomes, but not at the plasma membrane. Once internalized, lowaffinity agonists such as adrenaline dissociate rapidly from β2-adrenoceptors. 32 This event is followed by arrestin dissociation, because the tethering of high-affinity arrestin needs agonist binding, in addition to GRK site phosphorylation. The rates of endocytosis and recycling control the level of β2-adrenoceptors in the plasma membrane. Class-A GPCRs have higher affinity for arrestin-3 (or β-arrestin-2) than arrestin-2 (or β-arrestin-1). They internalize without arrestin and recycle rapidly to the plasma membrane. On the other hand, class-B GPCRs co-internalize with arrestin-2 or -3 and recycle slowly, often undergoing intracellular degradation. Once liganded, thyrotropin-releasing hormone receptor, neither class-A nor -B GPCR, is rapidly 31. Phosphorylation of GPCRs provokes a slight decrease in receptor activity. Above all, it also enhances GPCR affinity for arrestin. Arrestin binding can inactivate receptors by preventing their coupling to heterotrimeric G proteins triggered by agonist binding. In Drosophila, retinal degeneration-C (RDgC) is an unusual Ser-Thr phosphatase required for rhodopsin dephosphorylation [821] . Unlike vertebrate opsin, most invertebrate photopigments are not bleached after light activation, but can be photoconverted between the rhodopsin form and a thermally stable, active metarhodopsin form. 32. Agonists may be removed quickly from β2-adrenoceptors, especially in synapses, and GPCRs must be rapidly resensitized for appropriate functioning. β2-Adrenergic receptors contain phosphorylation sites for both GRK (Ser355 and Ser356) and PKA (Ser262) [823] . The concentration of agonist and rate constants of phosphorylation and dephosphorylation define the level of PKA-and GRK site phosphorylation. Dephosphorylation of the PKA site does not necessitate endocytosis, as it happens at the plasma membrane. Dephosphorylation of GRK sites occurs with a phase lag with respect to that of PKA site (possibly due to time necessary for phosphatase recruitment) and proceeds slowly. The rapid phase of resensitization (0-5 min) does not depend on dephosphorylation at the plasma membrane, but β-arrestin dissociation upon agonist stimulation of adenylate cyclase [823] . Rapid restoration of adenylate cyclase activity after removal of agonist can explain the fast dissociation of β-arrestin from membrane-bound β2ARs in association with recycling of the receptor to the plasma membrane. Moreover, the pattern of phosphatase inhibitor effects is much more consistent with PP1, rather than PP2 in PKA and GRK site dephosphorylation [823] . phosphorylated at numerous sites by GRKs, but not PKA enzyme. It then internalizes with arrestins-2 and -3. It can be dephosphorylated at the plasma membrane or in endosomes, once its hormonal agonist is removed. β2-Adrenergic receptor is slowly phosphorylated by both PKA and GRK kinases. The latter recruits arrestin, but internalizes without it. On the other hand, thyrotropin-releasing hormone receptor co-internalizes with arrestin. Both receptors are dephosphorylated following agonist removal, but the latter is dephosphorylated much more rapidly at the plasma membrane. Resensitization of most GPCRs comprises arrestin dissociation, receptor dephosphorylation, and recycling to the plasma membrane. Endocytosis often enables action of protein kinase-A, whereas GRKs mostly intervene at the plasma membrane. The GPCR cytoplasmic tails support arrestin linkage, but GPCR phosphorylation and dephosphorylation by GRKs and GRPs, respectively, depend on regions outside the cytoplasmic tail, but not their C-termini [824]. G-protein-coupled receptors stimulate heterotrimeric guanine nucleotide-binding protein and/or β-arrestins to regulate the intracellular concentration of various second messengers. Signaling is terminated by receptor desensitization and degradation of second messengers. Four arrestin isoforms exist: arrestins-1 and -4 that regulate opsins (GPCRs of photoreceptor cells of the retina) and arrestins-2 and -3 (or β-arrestins-1 and -2, respectively. 33 Ubiquitous β-arrestins control the activity of most G-protein-coupled receptors. 34 β-Arrestins were originally defined as terminators of GPCR signaling. They actually mediate desensitization and endocytosis of GPCRs and other types of receptors and molecules. 35 However, β-arrestins also act as signal transducers. They indeed serve as signaling adaptors for GPCRs and other types of receptors. β-Arrestin-biased agonism is related to GPCR signaling after ligand binding via β-arrestins rather than subunits of heterotrimeric G proteins. 33 . Some members of the arrestin family of adaptor proteins are called β-arrestins because they were discovered as β2-adrenoceptor-binding proteins. 34. β-Arrestins associate with β2-adrenergic receptor, angiotensin-2 type-1A receptor (AT 1A ), arginine vasopressin receptor (V 2 [AVPR2]), peptidase-activated receptor-2, and parathyroid hormone receptor, to activate extracellular signal-regulated protein kinases (Vol. 4 -Chap. 5. Mitogen-Activated Protein Kinase Modules). G-Protein-coupled receptor kinases are required, at least, for both AT 1A and V 2 receptors. 35. β-Arrestins act on receptor Tyr kinases, such as insulin-like growth factor-1 receptor. In addition, β-arrestins are implicated in endocytosis of VE-cadherin. β-Arrestin-2 also binds to low-density lipoprotein receptor and enhances LDLR endocytosis [825] . β-Arrestin-2 links to type-3 transforming growth factor-β receptor (TβR3; a.k.a. β-glycan) and triggers the receptor phosphorylation by type-2 TGFβ receptor kinase (TβR2), thereby downregulating TGFβ signaling [826] . Mechanical stretch induces β-arrestin-biased signaling from angiotensin-2 receptors AT 1 in the absence of ligand or G-protein activation to promote cell survival [827] . In cardiomyocytes, acute variations in mechanical stress trigger an AT 1mediated conformational change of β-arrestin similar to that caused by a ligand that primes a β-arrestin-associated process. Phosphorylated β-arrestins pertain to 2 classes: (1) class-A β-arrestin binders that transiently link GPCRs only at the plasma membrane, as they are deubiquitinated, and (2) class-B β-arrestin binders that form stable β-arrestin-GPCR complexes connected to endocytic vesicles. Ligand-stimulated ubiquitination of β-arrestins governs the stability of receptorβ-arrestin interactions. Ubiquitination of β-arrestins tunes signal strength and localization, as ubiquitination serves to further modify the active conformation of β-arrestin to recruit endocytic and signaling mediators among members of the βarrestin interactome. β-Arrestin ubiquitination by DM2 ubiquitin ligase causes rapid endocytosis of β2-adrenergic receptor. Deubiquitinase ubiquitin-specific peptidase USP33 binds βarrestin-2. Enzymes USP33 and DM2 favor the stability or lability of GPCR-βarrestin complex, respectively, thereby controlling the longevity and subcellular localization of signalosomes. β2-Adrenoceptors promote a conformational change of β-arrestin for deubiquitinase binding, hence destabilizing β2AR-β-arrestin binding and lowering ERK activation [828] . Conversely, ubiquitination of β-arrestin by DM2 promotes β-arrestin-dependent signaling of activated β2-adrenoceptors. On the other hand, V 2 vasopressin receptor elicits a distinct β-arrestin conformation that favors the dissociation of deubiquitinase, thereby stabilizating β-arrestin-receptor complexes and increasing ERK activation. In summary, DM2 ligase directs endocytosis and signaling, whereas deubiquitinase USP33 causes signal termination. β-Arrestins help to restore G-protein-coupled receptors to an inactive state after stimulation, although they can also have positive signaling roles. β-Arrestin-1 and -2 desensitize certain plasmalemmal receptors in conjunction with G-protein-coupled receptor kinases (GRK). 36 Agonist-promoted desensitization indeed comprises phosphorylation of the receptor by a GRK that promotes the binding of β-arrestin that partially uncouple GPCR from its effectors. This event can lead to GPCR endocytosis, with either a recycling to the cell surface or degradation. 36. β-Arrestins bind to activated G-protein-coupled receptors after receptor phosphorylation by G-protein-coupled receptor kinases. β-Arrestins recruit phosphodiesterases to activated Gs-coupled β2-adrenergic receptors to limit signaling. β-Arrestins also stop Gq-protein-coupled receptor signaling. In particular, β-arrestins regulate signaling primed by Gq-coupled M 1 muscarinic receptors by limiting the production of diacylglycerol and enhancing the degradation rate of this effector [829] . β-Arrestins indeed favor conversion of diacylglycerol into phosphatidic acid and interact with diacylglycerol kinases that degrade diacylglycerol. β-Arrestins also serve as endocytic and signaling adaptors. β-Arrestins indeed intervene as scaffolds for intracellular assembly of signaling complexes. Dissociation of β-arrestins allows recycling of G-protein-coupled receptors via Rab-dependent mechanisms. β-Arrestins target active kinases to specific locations within the cell and can regulate the lifetime of G-protein-coupled receptors in endosomes [830] . G-protein-coupled receptor kinases and β-arrestins switch off G-protein-dependent signaling, but transduce another kind of signaling from receptors, such as AT 1A angiotensin-2 receptor. 37 Such a signaling causes positive inotropy and lusitropy (Vols. 5 -Chap. 6. Heart Wall and 6 -Chap. 3. Cardiovascular Physiology), possibly either by regulating the cytosolic Ca ++ concentration or Ca ++ sensitivity of troponin and myosin-binding protein-C [831]. β2-Adrenergic receptors cooperate with Gi protein and β-arrestin in the cardiomyocyte, in addition to major β1-adrenergic signaling, to stimulate Ca V 1.2 channels and increase the intracellular Ca ++ concentration. On the other hand, β2adrenergic receptor stimulation via the Gs-PKA signaling inhibits TNFα-induced NFκB activation by stabilizing IκBα via β-arrestin-2. β-Arrestin-2 thus has an opposite role on NFκB signaling. The association of the GPCR Smoothened of the morphogen sonic Hedgehog (Sect. 10.2) with β-arrestin-2 and its phosphorylation by G-protein-coupled receptor kinase-2 promote Smoothened endocytosis via clathrin-coated pits [832] . 37. This signaling is thus carried out independently from the Gα q -PKC pathway. β-Arrestin interacts with the receptor and prevents the coupling between the receptor and G proteins. After binding of Wnt morphogens to Frizzled receptors (Sects. 7.13.22 and 10.3), cytoplasmic Disheveled is recruited to the plasma membrane and phosphorylated. The adaptor β-arrestin-2, also recruited by Frizzled-4, binds to phosphorylated Disheveled and allows Frizzled-4 endocytosis [833]. The bioactive phospholipid lysophosphatidic acid binds to cognate G-proteincoupled receptors (Sect. 7.13.35) to prime the signaling cascades and activate several transcription factors such as nuclear factor-κB (Vol. 4 -Chap. 9. Other Major Signaling Mediators). β-Arrestin-2 (but not β-arrestin-1) links to caspase-recruiting domain and membrane-associated guanylate kinase homolog domain-containing protein CARMA3 that is recruited to LPA receptors for NFκB activation and interleukin-6 production [834] . Scaffold protein CARMA3 associates with BCL10 and MALT1 to activate IκB kinase that target inhibitor of NFκB IκBα. Receptors of LPA trigger the Gq-PKC signaling with different post-translational modification of targets that can then serves as activators or inhibitors. Some GPCR-interacting proteins connect to specific GPCRs upon agonist binding to GPCRs to prime particular receptor signaling as mediators, whereas other interactors associate with receptors independently of agonists, thus acting as modulators, but not mediators. Transfer of and signaling from GPCRs depend on the context. Activity of G-protein-coupled receptors can be regulated by interactions between GPCRs and not only kinases, phosphatases, arrestins, and other receptor types, but also receptor-selective, cytoplasmic or transmembrane, proteic partners (Tables 7.23 and 7.24). G-Protein-coupled receptor-interacting proteins, the production of which differs according to the cell type, contribute to fine-tuning of GPCR function. Partners of GPCRs can: (1) regulate GPCR transport, as they can be required for exocytosis of their associated GPCRs down to the plasma membrane or sort endocytosis routes between recycling back to the plasma membrane or lysosomal degradation; (2) anchor GPCRs in suitable subcellular regions; (3) organize receptor signaling via G proteins; (4) act as scaffolds to modulate G-protein signaling, as they enhance signaling by tethering effectors in the vicinity of activated receptors or lower signaling intensity and/or duration either by impeding receptor-G-protein interactions or by Table 7 .23. G-Protein-coupled receptor interactors (modulators of GPCR signaling), excluding G proteins, GPCR kinases, arrestins, and regulators of G-protein signaling (Part 1; Source: [835] ; AKAP: A-kinase anchor protein [AKAP5 and AKAP12 are also denoted AKAP79 and AKAP250, respectively]; AR: adrenergic receptor; AT 1 : angiotensin-2 receptor; D 2 : dopamine receptor; DOR, KOR, MOR: δ-, κ-, μ opioid receptor; Fz: Frizzled; GABA: gamma-aminobutyric acid; JaK: Janus kinase; LPAR: lysophosphatidic acid receptor; MAGI: membrane-associated guanylate kinase; mGluR: metabotropic glutamate receptor; MT: melatonin receptor; MuPP: multiple PDZ domain-containing protein; PKA/C: protein kinase-A/C; PAFR: platelet-activating factor receptor; PTH1R: parathyroid hormone-1 receptor; STAT: signal transducers and activators of transcription; V 2 R: type-2 vomeronasal receptor). GPCR Action Among GPCR-associated proteins that enhance the efficiency of some G-proteinmediated signaling axes, members of the A-kinase anchoring protein family (AKAP) connect to β-adrenergic receptors and protein kinase-A to raise PKA-mediated phosphorylation of signaling mediators, such as β2AR and its effectors. On the other hand, GPCR interactors can reduce G-protein-mediated signaling. In addition to arrestins, spinophilin interacts with some GPCRs, such as dopamine, adrenergic, and muscarinic acetylcholine receptors and several regulators of G- Table 7 .24. G-Protein-coupled receptor interactors, excluding G proteins, GPCR kinases, arrestins, and regulators of G-protein signaling (Part 2; Source: [835] ; AR: adrenergic receptor; D 2 : dopamine receptor; DOR, KOR, MOR: δ-, κ-, μ-opioid receptor; LPAR: lysophosphatidic acid receptor; mAChR: muscarinic acetylcholine receptor; MCHR: melaninconcentrating hormone receptor; mGluR: metabotropic glutamate receptor; MT: melatonin receptor; Ncdn: neurochondrin; NHERF: Na + -H + exchange regulatory factor; P2Y: nucleotide receptor; PKA/C: protein kinase-A/C; PI3KR1/2: PI3K regulatory subunit (P85α/β); Ppl: periplakin [a.k.a. cornified envelope precursor protein]; PPP1R9b: protein phosphatase-1 regulatory (inhibitory) subunit-9B [a.k.a. PPP1R6, PPP1R9, neurabin-2, and spinophilin]; PTH1R: parathyroid hormone-1 receptor; RhoGEFi: Rho guanine nucleotide-exchange factor [i = 11, 12; a.k.a. ArhGEFi]; SSTR: somatostatin receptor). Reduction of G-and arrestin-mediated signaling α2AR Reduction of GPCR-mediated Ca ++ signaling mAChR Reduction of GPCR-mediated Ca ++ signaling protein signaling that act as GαGAP and can also link to some GPCRs to reduce the intensity and duration of GPCR-stimulated G-protein signaling. Calmodulin associates with Ca ++ and dopamine, metabotropic glutamate, and serotonin, as well as other receptors to lower G-protein coupling, hence directing a inhibitory feedback to restrain GPCR-initiated G-protein signaling. Nonetheless, G-protein-independent signaling can be potentiated by GPCR-calmodulin interactions. For example, the calmodulin-5HT 2C complex promotes arrestin-mediated signal transduction. Periplakin and neurochondrin, the former tethering to μ-type opioid receptor (MOR) and both to melanin-concentrating hormone receptor MCHR1 as well as few other GPCRs, attenuate G-protein-mediated signaling. Once synthesized, GPCRs take the exocytosis route to localize into the plasma membrane. After agonist stimulation, most GPCRs are internalized into endosomes Table 7 .25. Regulators of GPCR trafficking and/or ligand binding, excluding GPCR kinases, arrestins, and regulators of G protein signaling proteins (Part 1; Source: [835] ; AR: adrenergic receptor; AT: angiotensin-2 receptor; ATBP50: 50-kDa AT 2 receptor binding protein; CNR: cannabinoid receptor; D 1(2) : D1 (or D2) dopamine receptor; DOR, KOR, MOR: δ-, κ-, μ opioid receptor; DRIP78: 78-kDa dopamine receptor-interacting protein; EP: prostaglandin receptor; GABA: gamma-aminobutyric acid; GASP: GPCR-associated sorting protein; GEC: glandular epithelial cell protein; M10: M10 family members of the major histocompatibility complex class 1b; MAGI: membrane-associated guanylate kinase, WW and PDZ domaincontaining protein; mGluR: metabotropic glutamate receptor; MPP: MAGUK p55 subfamily member; MC2R: melanocortin receptor-2; MRAP: MC2R-accessory protein; MuPP1: multiple PDZ domain protein; NHERF: Na + -H + exchange regulatory factor; NINA: neither inactivation nor afterpotential protein; OdR: odorant response abnormal protein; PICK1: protein interacting with C kinase; PSD: postsynaptic density protein; SSTR: somatostatin receptor; V 2 R: type-2 vomeronasal receptor). Certain GPCR-interacting proteins regulate GPCR folding after translation before exocytosis as well as transport to and insertion into the plasma membrane. 38 38. Ran-binding protein RanBP2, dynein light-chain component T-complex testis-specific protein DynLT1 (a.k.a. TCTex1), glandular epithelial cell protein GEC1 (a.k.a. GABA A receptor-associated protein-like protein GABARAPL1), receptor of activated protein kinase RACK1 (a.k.a. guanine nucleotide-binding-like protein GNB2L1), dopamine receptorinteracting protein DRIP78 (a.k.a. heat shock protein HSP40 [DnaJ] molecular chaperone Other GPCR interactors influence post-endocytic transfer of GPCRs in response to agonist stimulation in a more receptor-selective manner than GRKs and arrestins. 39 Some GPCR interactors, such as Homer proteins and Disc large homologs, control GPCR anchoring to appropriate regions of the plasma membrane. DnaJC14), AT 2 -binding protein ATBP50, and ubiquitin specific-processing peptidase USP4 link to their corresponding GPCRs to enhance their transfer [835] . 39. GPCR-associated sorting protein GASP1 promotes endocytosis toward lysosomes of δtype opioid receptors (DOR), D 2 dopamine receptor, and CB 1 cannabinoid receptor [835] . On the other hand, sodium-hydrogen exchanger-regulatory factor NHERF1 promotes the recycling of β2-adrenoceptors and κ opioid receptors (KOR). Ligand-gated ion channels Some GPCR interactors influence agonist selectivity of their corresponding GPCRs. Transmembrane receptor activity-modifying proteins RAMP1 to RAMP3 tether to calcitonin receptor-like receptor to construct functional receptor for calcitonin gene-related peptide (RAMP1-CRLR) and adrenomedullin (RAMP2-CRLR and RAMP3-CRLR) as well as calcitonin receptor to form amylin receptors [835]. G-protein-coupled receptors represent drug targets. Handling of GPCR activation then allows design of agonist or antagonist ligands, in addition to signaling modeling. Muscarinic acetylcholine receptors (M 1 -M 5 or M1R-M5R) pertain to class-A GPCR receptors. Selective enhancers of acetylcholine binding and action exist for various receptor subtypes [5] . Main guanine nucleotide-binding (G) proteins targeted by muscarinic receptors are given in Table 7 .27. Muscarinic receptors M 1 and M 2 couple voltage sensing to acetylcholine binding. Conformational changes are associated with charge motions. 40 Ligand binding of GPCRs can then be modulated by voltage, amplifying or attenuating signals. Effects of muscarinic receptor subtypes are given in Table 7 .28. 40 . Voltage is sensed not only by voltage-gated ion channels, but also ion transporters, such as sodium-glucose cotransporter, voltage-dependent phosphoinositide phosphatase, and glutamate G-protein-coupled receptors mGlu 1 and mGluR 3 . The activation of muscarinic receptors in cardiomyocytes depends on transsarcolemmal potential. Depolarization reversibly induces changes in affinity state of the receptor [838] . Depolarization raises and reduces the affinity of M 1 and M 2 receptors, respectively [839]. The actin depolymerizing factor-cofilin complex regulates actin-dependent vesicular transport of acetylcholine receptors to the postsynaptic membrane and contributes to membrane insertion, formation, and maintenance of muscarinic receptor clusters [840] . Scaffold 14-3-3 that sequesters the ADF-cofilin complex attenuates the synaptic insertion of muscarinic receptors and clustering. On the other hand, 2 counteracting nerve-derived factors, agrin and acetylcholine, regulate the redistribution of muscarinic receptors on the sarcolemma at neuromuscular junctions. Agrin activates muscle-specific Tyr kinase (MuSK) that favors muscarinic receptor clustering on the postsynaptic membrane, whereas acetylcholine disperses extrasynaptic clusters of muscarinic receptors. The agrin-MuSK signaling involves P21-activated kinase, an activator of LIMK kinases that phosphorylates (inactivates) the ADF-cofilin complex. Protein 14-3-3γ colocalizes and interacts with MuSK at neuromuscular junctions. The M 1 subtype of muscarinic (acetylcholine) receptors is selectively distributed in the forebrain, where it intervenes in cognition and memory. It can mediate learning and memory via an indirect mechanism, i.e., stimulation of the prefrontal cortex. It also supports the synaptic transmission and excitatory postsynaptic potential in postganglionic nerves of autonomic ganglia. The M 1 receptor is randomly distributed over the plasma membrane [841] . At any time, about 30% of the receptors are dimers. (Oligomers M 1 are not detected.) Interconversion between M 1 monomers and dimers occurs on a time scale of seconds. Receptor M 1 transmits signal from the vagus nerves and cause a bronchoconstriction. In the digestive tract, M 1 provokes the secretion of salivary glands and gastric acid in the stomach. In the heart, M 2 receptor: (1) diminishes the speed of depolarization in the sinoatrial node (the natural pacemaker), thereby slowing the cardiac frequency, and (2) reduces the contractile force of atriomyocytes, but have no effect on that of ventriculomyocytes. Muscarinic M 2 and M 3 receptors are coexpressed in many types of smooth muscle cells, such as those in walls of the gastrointestinal tract, bladder, blood vessels, and airways. The density of M 2 receptors is greater than that of M 3 receptors (relative density ≥4:1) [842] . In airway smooth muscle cells, activated Gi-coupled M 2 receptor inhibits βadrenergic-mediated relaxation; activated Gq-coupled M 3 receptor initiates their contraction. In smooth muscle cells, M 2 receptor inhibits AC5 and AC6 adenylate cyclases via Gα i3 , whereas M 3 activates AC5 and AC6 via Gβγ [843]. Muscarinic M 3 receptor localizes to smooth muscle cells of the wall of blood vessels and respiratory conduits. This Gq-coupled receptor generates an increase in intracellular calcium concentration, thereby priming a contraction of smooth muscle cells (vaso-and bronchoconstriction). 41 However, in the vasculature, activated M 3 on vascular endothelial cells augment the synthesis of nitric oxide that targets adjacent vascular smooth muscle cells and causes their relaxation. Therefore, nitric oxide modulates the direct action of acetylcholine on smooth muscle cells. Functioning of pancreatic β cell is regulated by many hormones and neurotransmitters. Most of them act on specific G-protein-coupled receptors. Muscarinic receptor M 3 (or M3R) is linked to parasympathetic nerves that innervate the endocrine pancreas. In pancreatic β cells, M 3 receptor leads to the stimulation of G-protein subunits of the Gα q family and promotes glucose-stimulated insulin secretion. Subunit Gq are controlled by regulators of G-protein signaling. Protein RGS4, the most abundant RGS subtype synthesized in β cells, is a potent inhibitor of M 3 receptors [844] . Similarly to all GPCRs after agonist stimulation, the activity of M 3 is regulated via hyperphosphorylation by diverse protein kinases (GPCR kinases [GRK] and casein kinases CK1α and CK2), hence phosphorylation-dependent receptor endocytosis and β-arrestin recruitment. Additional arrestin-primed phosphorylation of M P 3 by protein kinase-D1 enables the sustained phase of glucose-dependent insulin release, independently of heterotrimeric G proteins [845] . 42 In the hippocampus, M 3 participates in learning and memory [846] . These cognition processes rely on M 3 phosphorylation, receptor endocytosis, and arrestin recruitment. Muscarinic acetylcholine receptors regulate dopaminergic neurotransmission. Receptor M 4 is coexpressed with D 1 dopamine receptor in a specific subset of striatal projection neurons. Activated M 4 receptor in the striatum inhibits D 1 -induced locomotor stimulation (in mice) [847] . Ca ++ ions from the sarcoplasmic reticulum. 42. Two phases of insulin release -transient and sustained -result from feeding. The early phase of insulin release depends on activation by Gq of calcium influx and protein kinase-C. Receptor M 5 is the sole mediator of ACh-induced vasodilation in the cerebral vasculature [848] . It is also involved in long-term potentiation at the hippocampal mossy fiber-CA3 synapse and hippocampal-dependent behavioral cognition. Adenosine is an ubiquitous, multifunctional purine nucleoside that targets numerous enzymes, such as protein kinases and adenylate cyclases. 43 Intracellular adenosine can be formed by either dephosphorylation of AMP by 5 -nucleotidase or by hydrolysis of S adenosylhomocysteine. Extracellular adenosine can be produced from released adenine nucleotides by a cascade of ectonucleotidases such as ecto-5 -nucleotidase (NT5E or CD73), which is highly expressed in microglial cells, and ectonucleoside triphosphate diphosphohydrolase ENTPD1 (or CD39) [849] . In the central nervous system, the extracellular concentration of adenosine is controlled by astrocytes. Glial cells and neurons release ATP, a neurotransmitter that activates its P2X and P2Y receptors. Vesicular ATP release from astrocytes (but not from neurons) by exocytosis or through equilibrative nucleoside transporters, possibly pannexin-1 hemichannels, and Maxi anion channels, is a source of extracellular synaptic adenosine via ectonucleotidase action on ATP [849] . Adenosine thus influences synaptic transmission. Endocrine cells also secrete ATP, even without releasing stored hormone. Adenosine receptors constitute a family of nucleoside receptors (P1 receptors; Table 7 .29). 44 Adenosine activates 4 receptor types (A 1 , A 2A , A 2B , and A 3 ). Gi-Coupled receptors A 1 and A 3 can also be activated by inosine. These adenosine receptors inhibit many intracellular ATP-using enzymes such as adenylate cyclases [5] . Adenosine is a cerebral vasodilator. Smooth muscle cells of cerebral arteries synthesize A 1 , A 2A , A 2B , and A 3 receptors [850] . Adenosine mainly binds to A 2A and A 2B and generates superoxide using NADPH oxidase and mitochondria. 43 . Purines adenine and guanine are synthesized in vivo via inosine monophosphate (IMP), which is laso named inosinic acid and inosinate. The latter is also converted into nucleotides, i.e., bases attached to ribose 5-phosphate: (1) adenosine monophosphate (AMP; a.k.a. adenylic acid and adenylate) by adenylsuccinate synthetase and adenylosuccinate lyase and (2) guanosine monophosphate (GMP; a.k.a. guanidylic acid, guanylic acid, and guanylate) by IMP dehydrogenase and GMP synthase. Conversely, AMP can be converted into IMP by myoadenylate deaminase. Adenosine can be transformed into inosine via adenosine deaminase and AMP via adenosine kinase. De novo synthesis of adenosine does not exist. 44. The P1 receptor class is associated with a nucleoside. The term "nucleoside receptor" to define a P1 family member is more appropriate then purinergic receptor. Members of the family of nucleotide receptors (P2) link to both purines and pyrimidines. As both purine and pyrimidine species are nucleotides, the P2 family is better defined as the class of "nucleotide receptor." Nevertheless, the noun "purinergic" should be kept in mind to explain the aliases P1 and P2. Adenosine receptors A 1 and A 3 stimulate K + channels, reduce transient activity of voltage-dependent Ca ++ channels, and inhibit cAMP formation. Whereas A 2A receptor couples to members of the Gs family (e.g., Gα olf in striatal neurons), A 2B receptor associates with many G-protein subunit types, such as Gs, Gq, and G12 [849] . Adenosine is approximately equipotent on A 1 , A 2A , and A 3 receptors. The A 2B receptor may require higher agonist concentrations. Adenosine A 1 receptor is ubiquitous (Table 7 .30). The A 1 receptor attenuates stimulatory actions of catecholamines on β-adrenergic receptors, reduces lipolysis in adipose tissue and urine production, and impedes neuronal activity [852] . Activated A 1 receptor not only decreases cAMP synthesis, but also stimulates phospholipase-C. Adenosine receptor A 1 contributes to sleep regulation, as it is upregulated in cortical and subcortical brain regions, especially in the orbitofrontal cortex after prolonged wakefulness [853] . It hence promotes sleep and inhibits wakefulness via cholinergic neurons, in opposition with xanthine, an adenosine antagonist in coffee and tea. The activity of basal forebrain (substantia innominata) and mesopontine (laterodorsal tegmental nucleus) cholinergic neurons actually bear an inhibitory control by endogenous adenosine in cats [854] . The A 1 receptor in the nucleus of the solitary tract is involved in the baroreflex (Vols. 2 -Chap. 1. Remote Control Cells and 6 -Chap. 3. Cardiovascular Physiology). Its activity inhibits glutamatergic transmission in the nucleus of the solitary tract [855] . This receptor differentially inhibits and resets the baroreflex response of preganglionic adrenal, renal, and lumbar sympathetic nerve. Glial cells that interact with neurons and blood vessels control adenosine that has a neuroprotective role in the central nervous system. Microglial cells, astrocytes, endothelial cells, oligodendrocytes, and neurons produce signals to manage inflammation, hence entry of cells of the innate and adaptive immune system in the brain. These cells release arachidonic acid metabolites, nitric oxide, cytokines, and chemokines as well as ATP and adenosine. The A 1 receptor on microglial cells reduce excessive immune activation of microglial cells [849] . It also stimulates oligodendrocyte migration. The A 1 receptor on neurons, especially at nerve terminals, contributes to the dampening of neuronal activity mediated by adenosine generated from ATP released from astrocytes [849] . In nerve endings, it may preferentially signal via Go proteins to inhibit calcium channels. In neuron bodies and dendrites, it may preferentially regulate potassium channels via Gi proteins. High-affinity A 2A and low-affinity A 2B receptors are coupled to stimulatory subunit Gα s of heterotrimeric G protein, thereby raising intracellular cAMP concentration (Table 7 .31). Activated A 2A can protect against tissue injury and act as an anti-inflammatory agent [856] . Human A 2A receptor has a binding site that differ in position and orientation with respect to other GPCRs [857] . Extracellular loops in collaboration with the helical core allows ligand recognition. In the brain, A 2A receptor is synthesized at high levels in striatal neurons and at low levels in other neurons and glial cells [849] . The A 2A production in glial cells rises after brain insult. In astrocytes, activated A 2A by extracellular adenosine increases cell proliferation, but impedes NOS2 synthesis. In microglial cells, activated A 2A facilitates the release of cytokines and prostaglandin-E2, as it boosts the activity of cyclooxygenase-2, as well as that of nitric oxide synthase and production of nerve growth factor [849] . Activated A 2A in the nucleus of the solitary tract in the brainstem increases activity of preganglionic adrenal sympathetic nerves, but decreases that of renal sympathetic nerves [858] . The A 2B receptor can couple with Gq and Gs on astrocytes. It is responsible for the adenosine-induced stimulation of interleukin-6 from astrocytes. Increased neural activity is associated with cerebral vasodilation. Neural activation-associated pial arteriolar dilation involves interactions between A 2 and inward rectifier K IR channels [859] . Large-conductance, Ca ++ -activated BK channel (K Ca 1) participates in the process within the glia limitans. The A 2 receptor provokes the synthesis by adenylate cyclase of cAMP that primes the phosphorylation of K + channels by protein kinase A. In human dermal microvascular endothelial cells, activation of A 2B , but not A 2A , promotes angiogenesis. On the other hand, activated A 2A , but not A 2B , promotes angiogenesis in endothelial cells of human umbilical veins and lung microvasculature. Endothelial cells express A 2A , A 2B , or both. In the respiratory tract, A 2B receptors regulate chloride channels via cyclic adenosine monophosphate. Like the adenosine receptor A 1 , A 3 is coupled to inhibitory subunit Gα i of heterotrimeric G protein, thus inhibiting adenylate cyclase (Table 7 .30). It has a protective function during tissue ischemia. Adenosine receptor A 3 contributes to inhibition of neutrophil degranulation in tissue injury. In neutrophils, ATP acts on P2Y receptors in synergy with adenosine that binds to A 3 receptors to stimulate migration. Astrocytic A 3 receptor regulates chemokine release [849] . Activation of A 3 by adenosine protects astrocytes from death caused by hypoxia. Adenosine contributes to microglial migration caused by ATP predominantly via P2Y 12 receptors. Fluorescence resonance energy transfer-based assays can be used to assess the receptor-specific activation and deactivation kinetics of the different GPCR signaling steps, especially the activation kinetics of initial stages (GPCR-G-protein interaction that depends on the amount of available receptors and determines the activation Gprotein and other effectors [860] . Both A 2A and β1-adrenergic receptors couple to Gs protein, thereby stimulating adenylate cyclase that produces cAMP messenger. Times of A 2A receptor activation, A 2A -Gs interaction, and Gs activation following stimulation by A 2A or β1adrenoceptors are smaller than 40, 60, and 550 ms, respectively. Only a fraction of Gs proteins is activated when GPCR-Gs interaction is maximal. The rate of Gs activation limits the signaling initiation via Gs-coupled receptors. Activation time of Gs subunit is also similar to that of Gi stimulation after excitation by β1-adrenergic receptors. Three main events occur between GPCR-G-protein interaction and G-protein activation: GDP release from Gα, GTP binding, and conformational change. Release of GDP is rate-limiting in G-protein activation. Half-time of cAMP activation is about equal to 35 s, but activation quickly appears (a few ssconds) due to the existence of Gs-ACase complexes. In addition, although the activation kinetics via A 2A and β1-adrenergic receptors are similar, the deactivation kinetics differ. Termination of A 2A -Gs interaction and Gs deactivation (following A 2A deactivation with a time ∼2 s, which is similar to that of α2a-adrenoceptors) are significantly slower than that of β1-adrenoceptor signaling (termination of β1-adrenoceptor-Gi interaction ∼13 s). Termination of A 2A -Gs interaction is faster than Gs deactivation, which is linked to the activity of accessory proteins (activators and regulators of G-protein signaling) involved in GPCR signaling. Ligand washout is hence not limiting. Kinetics of A 2A recptor are given in Table 7 .32. In the vasculature, A 1 receptors as well as A 2A , A 2B , and A 3 are synthesized by smooth muscle cells [861] . The A 1 receptor causes contraction of vascular smooth muscle via phospholipase-C. It hinders vascular relaxation mediated by other adenosine receptor subtypes. The A 1 receptor upregulates PKCα, PKCβ1, PKCβ2, PKCγ, PKCε, and PKCζ isoforms (but not PKCδ and PKCμ) in a dose-dependent manner [862] . Activated A 1 tethers to Gi subtypes Gi1 to Gi3 or Go subunit, thereby inhibiting adenylate cyclase and decreasing cAMP concentration. Gs-Coupled A 2A and/or A 2B receptors are involved in adenosine-mediated vascular relaxation of coronary and aortic beds in mice [861] . Gi-Coupled A 3 receptor does not cause a vasoconstriction, as it is inhibited by A 2A receptor. The A 3 receptor mediates the cardioprotective effect of adenosine. In the central nervous system, adenosine, a potent vasodilator, intervenes in vasodilation during hypotension within the autoregulatory range of vascular autoregulation via both A 2A and A 2B receptors [863] . Hypoxia stimulates hypoxia-inducible HIF1α and HIF2α transcription factors that promote angiogenesis. 45 The HIF2α factor, but not HIF1α, regulates A 2A receptor in pulmonary endothelial cells to increase cell proliferation and migration as well as elicit tube formation [856] . Cardiomyocytes, in particular ventriculoyocytes, express the 4 adenosine receptor subtypes. Receptors A 1 and A 2A regulate myocardial oxygen consumption and coronary blood flow. Adenosine is released from the heart subjected to hypoxia or adrenergic stimulation. The A 1 receptor has a strong anti-adrenergic effect. Therefore, adenosine antagonizes adrenergic stimulation to avoid overstimulation. Stimulation of A 1 activates Gi subunit, releases the Gβγ dimer, and excites phospholipase-C and protein kinase-C [864]. In addition, activated Gi subunit counteracts effect of Gs subunits stimulated by catecholamines via adrenoceptors. The A 1 receptor is involved in the protection acquired during ischemic preconditioning against myocardial ischemia-reperfusion injury [865] . Stimulated A 1 re-45. In almost all cell types, HIF1α targets various genes, such as those that encode vascular endothelial growth factor and its receptors, glucose transporter-1, carbonic anhydrase-9 and -12, hexokinase-2, glucose phosphate isomerase, phosphofructokinase, aldolase-A and -C, glyceraldehyde 3-phosphate dehydrogenase, among others. On the other hand, HIF2α regulates a few genes in specific cell lines, such as those that encode erythropoietin, superoxide dismutase-1 and -2, glutathione peroxidase, catalase, frataxin, Octamer-4, insulin-like growth factor-binding protein IGFBP3, SRY-related HMG-box gene product Sox9, CBP and P300interacting transactivator with Glu/Asp-rich C-terminal domain CITED2. In most cell types, the gene set regulated by HIF2α overlaps with that of HIF1α [856] . presses the pacemaker cell function and decreases action potential conduction, thus reducing the heart rate [852] . It also lowers the atrial contractility. 46 Whereas the A 1 receptor reduces β1-adrenergic receptor-induced increase in contractility in rat ventriculomyocytes, the A 2A receptor enhances cardiac contractility via Gs and Ca ++ transient [868] . Cardiac A 1 and A 2A receptors use PKC and the cAMP-PKA pathway, respectively. Cardiac A 2A receptor acts like β1-adrenoceptor, as it also primes the cAMP-PKA pathway, but with smaller Ca ++ transient amplitude. The A 2A receptor, but not the A 2B receptor, counteracts the A 1 receptor. It indirectly enhances heart contractility, as it modulates the A 1 anti-adrenergic effect [869] . On the other hand, the A 2B receptor exerts a direct contractile effect, but does not alter β-adrenergic or A 1 anti-adrenergic effects. Activated A 2A receptor before reperfusion following coronary artery occlusion reduces infarct size and improves cardiac function, but activation must occur before or less than 1 h after reperfusion [870]. The A 2 receptor acts as a sensor of tissue damage. The A 2A receptor attenuates inflammation and tissue damage. The A 2B receptor abounds in vascular smooth muscle cells. It regulates the activity of the G-protein-coupled CXCR4 receptor that attracts vascular progenitor cells from bone marrow during tissue regeneration, avoiding excessive tissue growth after injury [871] . Chemokine CXCR4 receptor controls migration of inflammatory cells and platelet aggregation. It is thus involved in vascular repair and remodeling after injury. 47 Upregulation of CXCR4 in injured vessels is limited by A 2B receptors. In addition, adenosine inhibits smooth muscle cell proliferation via activated A 2B receptor. 46. Overexpression of A 1 causes supraventricular arrhythmias [866] . A strong A 1 overexpression also induces dilated cardiomyopathy [867] . 47. Chemokine CXCR4 receptor is expressed on the surface of macrophages, lymphocytes, hematopoietic stem cells, bone marrow stromal cells, megakaryocytes, and platelets. It is involved in the migration of hematopoietic progenitors and stem cells during cardio-and vasculogenesis and hematopoiesis. It is regulated by TNFα secreted especially by several bone marrow cell types. Nucleotide receptors of the P2Y family bind to both purine and pyrimidine nucleotides. 48 Purines include nucleic acid bases adenine and guanine 49 as well as hypoxanthine, xanthine, theobromine, caffeine, uric acid, and isoguanine. 50 Ubiquitous nucleoside and nucleotide receptors (or purine and pyrimidine receptors) include P1 receptors (A 1 -A 3 ) and P2 receptors (P2X and P2Y; Table 7 .33). P1 Receptors bind adenosine, P2 receptors extracellular adenosine and purine nucleosides and nucleotides [872] ( Fig. 7 .4). Ionotropic P2X receptors that encompass 7 known types (P2X 1 -P2X 7 ) are ion channels (Sect. 2.5.7). 51 Most P2X channels are cation-selective pores that discriminate more or less efficiently different cations, particularly Ca ++ ions. Many identified metabotropic, G-protein-coupled P2Y receptors (P2Y 1 -P2Y 14 ) are activated by ATP, ADP, UTP, UDP, and UDP glucose. The P2Y receptors differ according to their ligands (adenine vs. uracil-derived nucleotides as well as tri-vs. diphosphonucleotides), signaling mode, and cell expression (Tables 7.34 and 7.35). Calcium fluxes generated by P2Y receptors are larger for a given depolarization than equivalent amplitude hyperpolarization [873]. 48. The term "purinergic receptors" used to designate receptors of the P2Y family is inappropriate. In addition, receptors of the P1 family connect to purine nucleoside adenosine. The term "nucleotide receptors" to denote both members of the P1 and P2Y family of the GPCR superclass should be likewise avoided. 49. Double-stranded DNA relies on base pairing between nucleotide constituents. Purines adenine and guanine couple with pyrimidines thymine and cytosine, respectively. Whereas each DNA strand is composed of 4 bases -adenine, cytosine, guanine, and thymine -, single-stranded RNA contains adenine, cytosine, guanine, and uracil. 50. Purines are components of numerous molecules, such as: (1) purine nucleosides adenosine and guanosine; (2) adenylate (ATP, ADP, AMP, and cAMP) and guanylate (GTP, GDP, GMP, and cGMP) nucleotides; and (3) nucleotides constituents of enzymatic cofactors (coenzyme-A, flavin adenine dinucleotide [FAD], flavin mononucleotide [FMN] , and nicotinamide adenine dinucleotide phosphate [NADH] ). Pyrimidines include nucleobases cytosine, thymine, and uracil. Uridine is a pyrimidine nucleoside. Pyrimidine nucleotides encompass uridine mono-(or 5 -uridylic acid [UMP]), di-(UDP), and triphosphate (UTP). 51. Nucleotide receptors P2X are fast-acting, ligand-gated channels that are selective for calcium. When ATP binds to plasmalemmal P2X receptors, extracellular Ca ++ enters and activates signal transduction pathways. These homo-and hetero-oligomeric channels contain 2 transmembrane domains. Nucleotides can be stored and secreted upon adequate cell excitation to serve as messengers that bind to P2Y receptors. In the chromaffin granules of the adrenal medulla, ATP concentration reaches 100 mmol (cytoplasmic concentration 3-5 mmol) [874] . Dense granules of platelets contain about 500 mmol of ADP and ATP, as well as lower amounts of other nucleotides (adenine dinucleotides, GTP, and UTP). Following secretion and/or degranulation upon appropriate stimulation, 52 concentration of released nucleotides transiently reaches high levels. Nucleotide concentration is much higher near producing cell surface than in bulk extracellular fluid. 52. Change in blood flow pattern induces ATP release from endothelial cells. Release of ATP from human nasal epithelial cells increases upon mechanical stimulation or rising intracellular concentration of calcium ions. Corelease of ATP and nucleotidases from sympathetic nerves terminates the action of a neurotransmitter. Table 7 .34. Adenosine P1 and nucleotide P2Y receptors, their main targeted G proteins, and order of ligand potency (Sources: [736, 874] ). The P2X receptors are ATP-gated ion channels for Na + , K + , and Ca ++ ions. Some P2Y receptors couple exclusively to a single Gα-protein subunit, whereas others are dually coupled to different Gα-protein subunits (e.g., Gq/11 and Gi). Four subtypes of P1 receptors (A 1 , A 2A , A 2B , and A 3 ) exist. The P1/A 2 receptors are coupled to Gs subunit and stimulates adenylate cyclase, whereas P1/A 1 and P1/A 3 receptors couple to subunits of the Gi/o family and inhibit adenylate cyclase. The P2Y 5 receptor corresponds to LPA 6 receptor that primes the G13-Rho pathway. The P2Y 9 receptor (or LPA 4 ) is most closely related to P2Y 5 (both act as lipid and nucleotide receptors). Main transducer Potency order Adenosine receptors (preferentially) Gi/o, Gq/11 UTP > ATP P2Y 6 Gq/11 UDP UTP > ATP P2Y 11 Gs, Gq/11 ATP > UTP P2Y 12 Gi/o ADP ATP P2Y 13 Gs, Gi/o ADP ATP P2Y 14 Gi/o, Gq/11 UDP glucose (= UDP galactose) Extracellular nucleotides operate as auto-or paracrine mediators (Tables 7.36 and 7.37). Afterward, extracellular nucleotides are rapidly degraded by ubiquitous ectonucleotidases on the cell surface. 53 53. Two families of integral membrane ectonucleotidases exist: ectonucleotide diphosphohydrolases (ENPDH or CD39) and ecto-phosphodiesterase-nucleotide pyrophosphatase Inhibition of platelet aggregation Nucleotides exert their effects mainly via binding P2 receptors, although phosphorylation of plasmalemmal proteins by ectoprotein kinases constitutes an additional mechanism by which ATP can modulate cell function. 54 Nucleotide P2Y receptors can be partly characterized by order of potency of ligand binding. These receptors are activated by a set of nucleotides: adenosine diphosphate (ADP), adenosine triphosphate (ATP), uridine triphosphate (UTP), and uridine diphosphate-glucose ( UDP glucose; Table 7 .34). 55 The P2Y receptors can possess a main ligand, whereas other nucleotides behave as partial agonists. Nonetheless, (EPDNP) . The latter catalyze the cleavage of ATP into AMP and pyrophosphate, the conversion of cAMP into AMP, as well as several ATP-ADP, ADP-AMP, and AMP-adenosine reactions. Agent CD39 expressed by vascular endothelial cells hydrolyzes both ATP and ADP released from platelets. 54. Various plasmalemmal proteins are phosphorylated in their ectodomains by cell-surface protein kinase, the ectoprotein kinase. Protein CD36 (a.k.a. glycoprotein-3b and -4) is a class-B scavenger receptor that binds many ligands, such as collagen, thrombospondin, lipoproteins, oxidized low-density lipoproteins and phospholipids, as well as long-chain fatty acids. In platelets, cAMP-dependent ectoprotein kinase phosphorylates CD36 and causes platelet reactivity and adhesion to collagen, but reduces thrombospondin binding. Conversely, CD36 dephosphorylation transforms CD36 into a thrombospondin receptor [874] . 55. Nucleotide UDP is phosphorylated into UTP by uridine diphosphokinase. some nucleotide P2Y receptors can be activated almost equipotently by 2 nucleotide species. Others can be ligand-selective receptors, such as purinoceptors (e.g., P2Y 1 ) and pyrimidinoceptors (e.g., P2Y 6 ), for which ADP and ATP are either strong or only active ligands, or very weak stimulators or fully inactive, respectively. In addition, effect of a given nucleotide on a given nucleotide receptors P2Y can depend on mammalian species, as P2Y can require either full or partial agonist activation. Moreover, several types of P2Y receptors can colocalize in a given cell type. The P2Y receptors are coupled to distinct G proteins (Gq/11, Gi/o, and Gs; Table 7.34). They are then associated to phospholipase-C (Table 7 .35). They can stimulate adenylate cyclase via Gα s , although other pathways can be used (activation of some adenylate cyclase isoforms by Gβγ subunits, of other isoforms by Ca ++calmodulin or protein kinase-C, and mediation by prostaglandins). Conversely, they can inhibit adenylate cyclase via Gα i . The P2Y 1 receptor is linked to the RhoA-RoCK pathway (Vol. 4 -Chaps. 8. Guanosine Triphosphatases and Their Regulators and 4. Cytosolic Protein Ser/Thr Kinases) in addition to phospholipase-C. The P2Y 2 receptor is linked to PLCβ1 via Gα q/11 and to PLCβ3 via Gβγα i3 . In vascular endothelial and smooth muscle cells, activation of P2Y 2 receptors by ATP and UTP induces phosphorylation (activation) of extracellular signal-regulated protein kinases ERK1 and ERK2 downstream from phospholipase-C [874] . In rat cardiac fibroblasts as well as pressure-overloaded mouse cardiomyocytes, ATP stimulates P2Y 2 receptor and activates nuclear factors of activated T cells NFAT1 and NFAT3, thereby increasing the concentration of inducible NO synthase (NOS2) [875] . Enzyme NOS2 interacts with the P65 subunit (or RelA) of NFκB and causes its S-nitrosylation. 56 Subunit P65 SNO NFκB has a reduced transcriptional activity. Consequently, the density of type-1 angiotensin receptor (AT 1 ) falls. The P2Y receptors can also partner with ion channels. The P2Y 2 receptor links to Ca ++ -dependent Cl − , inward-rectifier K + , and Ca V 2.2 channels [874] . Receptor P2Y 6 can associate with Cl − channels. Via UDP, it can hamper the activity of K V 7.2 and K V 7.3 channels. In neurons and other excitable cells, GPCRs can modulate the activity of voltagegated ion channels, by mainly closing, or in certain circumstances opening or potentiating, various types of K + channels and voltage-gated Ca ++ and Na + channels, via G-protein subunits and protein kinases. Specific couplings between P2Y receptors and certain Ca ++ and K + channels occur in short time scales (O[100 ms]; Table 7.38) [876] . Cells of the superior cervical ganglion (SGC) are endowed with 2 main types of voltage-gated channels: (1) K V 7.2 and K V 7.3 (M-type K + channel) and (2) Ca V 2.2 (N-type Ca ++ ) channels. In SCG neurons, Gi/o-coupled P2Y 12 and P2Y 13 receptors close Ca V 2.2 channel. The P2Y 1 and P2Y 2 receptors act similarly to Gq/11-linked M 1 receptor. Both UDP and UTP nucleotides are strong agonists of P2Y 6 that provoke Ca V 2.2 channel closure [876]. Immunocytes release the auto-and paracrine controller ATP that activates P2X and P2Y receptors. Stimulated T-cell receptors provoke Ca ++ influx in the cytosol and then mitochondria, where it promotes ATP synthesis. Messenger ATP, a costimulatory signal to T lymphocytes, released from activated T lymphocytes through pannexin-1 hemichannels excites P2X receptors for a sustained mitogen-activated protein kinase signaling [877] . On the other hand, oxidized ATP, a P2X antagonist, impedes MAPK activation in stimulated T cells. Nucleotide signaling is also required for neutrophil activation. Activated neutrophils release ATP in response to exogenous stimuli such as formylated bacterial peptides and inflammatory mediators that stimulate Fcγ(Fc receptors for IgG), interleukin-8, C5a complement component, and leukotriene-B4 receptors. Stimulated formyl peptide receptors that colocalize with nucleotide P2Y 2 receptors 56. β-Arrestins enable the formation of the quaternary arrestin-P65-IκB-NOS2 complex. Nucleotides increase the apical permeability to Cl − in airway epithelial cells and enhance mucin secretion by goblet cells and submucosal gland secretion [874] . They then promote mucociliary clearance. 57 Nucleotides ATP and UTP not only augment formation of inositol phosphates and intracellular calcium concentration in human airway epithelial cells via P2Y 1 and P2Y 2 , but also regulate outwardly rectifying chloride channels via P2Y 2 receptors. Like muscarinic receptors, the activity of P2Y 1 receptors depends on the plasmalemmal potential. Nucleotide receptors participate in the regulation of the cardiac function, as P2Y receptors (especially P2Y 6 , P2Y 7 , P2Y 9 , and P2Y 14 ) reside in heart ( Fig. 7 .5). In blood vessel walls, nucleotide signaling controls the vascular tone, smooth muscle cell proliferation, and platelet aggregation. Different types of cells in the cardiovascular system selectively express nucleotide receptor types (Fig. 7.6 ). Contraction of vascular smooth muscle cells results from activated P2X 1 and several P2Y subtypes (P2Y 2 , P2Y 4 , and P2Y 6 ). 58 The P2X 1 receptor causes a rapid 57. In cystic fibrosis, CFTR mutations limit Cl − secretion and cause formation of underhydrated, viscous mucus that obstructs airways. Aerosolized UTP may be used to treat cystic fibrosis. 58. Contractile type of smooth muscle cells possess larger amounts of P2Y 4 and P2Y 6 , but a lower quantity of P2Y 2 , than synthetic type of smooth muscle cells. ). Messenger ATP, a cotransmitter of noradrenaline and neuropeptide-Y, released from perivascular sympathetic nerves binds receptors P2X 1 , P2X 2 , and P2X 4 of smooth muscle cells and induces vasoconstriction. Adenosine produced from ATP degradation binds to receptors A 1 (P1) of sympathetic nerves and inhibits transmitter release. Stressed endothelial cells release: (1) ATP, which binds receptors P2Y 1 (with a selective agonist action from ADP) and P2X 4 , and (2) UTP, a ligand for P2Y 2 . Both ATP and UTP trigger nitric oxide delivery to smooth muscle cells with subsequent vasodilation. influx of Ca ++ to initiate smooth muscle contraction. Gq/11-Coupled P2Y receptors activate phospholipase-C, thereby generating IP 3 , which mobilizes Ca ++ from its intracellular stores. Prolonged P2Y signaling can cause vasoconstriction, hypertension, vascular smooth muscle hypertrophy, and hyperplasia. However, GPCR-desensitizing GRKs and arrestins prevent prolonged and inappropriate GPCR-initiated signaling. 59 In mesenteric arteries, both GRK2 (but not GRK3, GRK5, or GRK6) and arrestin-2 (but not arrestin-3) regulate UTP-stimulated P2Y 2 on smooth muscle cells [880] . Endothelial cells express nucleotide receptors P2Y 1 , P2Y 2 , P2Y 4 , P2Y 6 , and P2Y 11 , 60 as well as P2X 1 , 61 to P2X 7 . 62 The P2X 4 receptor is the most abundant P2X receptor subtype in vascular endothelial cells. It contributes to ATP-and flowinduced Ca ++ influx in endothelial cells. The P2 receptors (particularly P2X 4 ) on endothelial cells bind ATP that triggers secretion of nitric oxide and causes vasodilation. The P2X receptors are implicated in autocrine loops in endothelial cells. Vascular smooth muscle cells abundantly produce P2X 1 , P2Y 2 , and P2Y 6 receptors. The P2 receptors also localize to the vessel wall adventitia. Perivascular adrenergic and cholinergic nerves have nucleotide receptors. Endothelial cells that ensure hemostasis release nucleotides, but ectonucleoside triphosphate diphosphohydrolase ENTPD1 63 on the surface of endothelial cells can degrade both anti-aggregatory ATP and pro-aggregatory ADP [874] . Further cleavage by nucleotidase generates adenosine that inhibits platelet aggregation via A 2A receptors and cAMP messenger. Moreover, ATP and ADP prime the release of antiaggregatory mediators prostacyclin and nitric oxide from endothelial cells, as they activate endothelial P2Y 1 and P2Y 2 receptors. Erythrocytes release ATP when the blood partial pressure of oxygen decreases. During exercise, when oxygen extraction by the myocardium is high, 64 the coronary blood flow rises. This augmentation is correlated with ATP concentration, as ATP 59. Among 7 GPCR kinases (GRK1-GRK7) and 4 arrestin (arrestin-1-arrestin-4) subtypes, GRK2, GRK3, GRK5, and GRK6, and arrestin-2 and -3 (i.e., β-arrestins) are ubiquitous. The GRK2 kinase is an important subtype for the contraction of vascular smooth muscle cells triggered by AT 1 , α1d-adrenergic, and ET A receptors. 60. The P2Y 4 receptor is slightly expressed in endothelial cells, whereas P2X level increases in blood of the coronary venous sinus (from 31.1 to 51.2 nmol) [883] . Adenine nucleotide ATP is one of the factors that control the coronary blood flow during exercise. It acts on microvascular endothelial cells to produce a vasodilation of upstream arterioles to match the oxygen delivery to myocardial oxygen consumption during exercise. Adenosine triphosphate released from erythrocytes is catabolized into ADP and AMP in coronary capillaries, where these products target nucleotide receptors such as P2Y 1 on the surface of endothelial cells [884] . Activated nucleotide receptors initiate a retrograde vasodilatory signal to upstream arterioles presumably via gap junctions to control coronary blood flow resistance using a feedback loop. The P2Y 13 receptor is synthesized in blood leukocytes. The P2Y 11 receptor participates in granulocytic differentiation. Agent ATP can trigger hematopoiesis via the P2Y 11 -Gs-ACase-cAMP-PKA pathway [874] . Major platelet activators include ADP, an auto-and paracrine regulator secreted by platelets, and thrombin, a peptidase of the coagulation cascade that triggers the release of ADP by platelets. Platelet activators interact with their respective GPCRs to prime and amplify platelet activation and aggregation. Binding of von Willebrand factor, collagen, and thrombin to their corresponding receptors on thrombocyte surface enhances the release of ADP and thromboxane-A2. Sustained platelet aggregation by ADP requires activation of both P2Y 1 and P2Y 12 receptors. Ligand ADP binds to: (1) Gq-coupled P2Y 1 receptor to cause cell shape change for optimal affinity for fibrinogen and von Willebrand factor, calcium mobilization, and initiation of reversible aggregation; 65 and (2) Gi-coupled P2Y 12 receptor to amplify the platelet aggregation via the ACase-cAMP pathway [885] . Metabotropic receptor P2Y 1 also mediates the release of nitric oxide caused by adenine nucleotides. In platelets, nucleotide (ADP) P2Y 12 receptor functions downstream from peptidase (thrombin)-activated PAR 1 and PAR 4 receptors to amplify the platelet response to thrombin, but can also operate in collagen-induced platelet activation and thrombin-PAR-independent pathways [886] . Furthermore, P2Y 12 signaling is more important than that based on PAR receptors in hemostasis, but not in arterial thrombosis for which PAR signaling is predominant. In addition, ionotropic receptor P2X 1 abounds in platelets. Agent ATP, a potent agonist of P2X 1 , triggers a rapid and quickly desensitized Ca ++ influx. 65 . The resulting platelet activation triggers a conformational change in α 2B β 3 -integrins (or glycoproteic complex GP2b-GP3a) that increases their affinity for fibrinogen and von Willenbrand factor. These ligands then bind to receptors to bridge adjacent platelets. The P2X 4 receptor improves survival of cardiomyocytes in a mouse model of cardiomyopathy [887] . Nucleotides intervene in synaptic transmission. In neurons of the central and peripheral nervous system, ATP colocalizes and is cosecreted with neurotransmitters, such as catecholamines and acetylcholine. The P2 receptors localize in every part of the nephron that regulates water (blood) volume and electrolyte concentration (Table 7 .39). Adiponectin receptors (Table 7 .40; Sect. 6.9.1) respond to adiponectin synthesized in adipose tissues (Vol. 2 -Chap. 3. Growth Factors). Unlike typical GPCRs, its C-and N-termini are extra-and intracellular, respectively [5] . Receptor AdipoR1 abounds in the skeletal muscle and liver, whereas AdipoR2 is predominantly produced in the liver. These receptors are produced by endothelial cells and cardiomyocytes. T-cadherin (Vol. 1 -Chap. 7. Plasma Membrane) may act as a coreceptor for mid-(Apn m ) and high-molecular-weight (Apn h ) adiponectin on endothelial and smooth muscle cells, but not low-molecular-weight trimeric (Apn l ) and globular ( G Apn) forms [889, 890] . Signaling via adiponectin receptors avoids G proteins and instead stimulates protein phosphorylation via AMP-activated protein kinase (Vol. 4 -Chap. 4. Cytosolic Protein Ser/Thr Kinases) and the mitogen-activated protein kinase module, possibly using Adaptor phosphoTyr interaction, PH and Leu zipper domain-containing protein APPL1. Both Ca ++ -calmodulin-dependent protein kinase kinase CaMKKβ and protein Ser/Thr kinase STK11 (or LKB1) are required for adiponectin-induced full AMPK activation. Adiponectin is an antidiabetic and anti-atherogenic adipokine. 66 It induces influx of extracellular Ca ++ via adiponectin receptor AdipoR1. Subsequent activation of Ca ++ -calmodulin-dependent protein kinase kinase-β, AMPK, and sirtuin-1. Both AMPK and SIRT1 are required for elevated expression and attenuated acetylation of peroxisome proliferator-activated receptor-γ coactivator PGC1α primed by the Apn-AdipoR1 axis. This pathway raises mitochondrial content and activity in myocytes [891] . It also heightens oxidative type-1 myofibers and oxidative stressdetoxifying enzymes in the skeletal muscle, thereby promoting insulin sensitization and exercise endurance. Two main groups of ubiquitous adrenergic receptors (or adrenoceptors) include αand β-adrenergic receptors (AR; Table 7 .41). Both groups bind both adrenaline and noradrenaline. These catecholamine messengers regulate diverse physiological systems. Adrenergic receptors are composed of 3 major types, each of which is further subdivided into 3 subtypes: (1) α1-adrenoceptors with α1a (or α1c), α1b, and α1d subtypes; (2) α2-adrenoceptors with α2a (or α2d), α2b, and α2c subtypes; and (3) β-adrenoceptors with β1, β2, and β3 subtypes. Each adrenoceptor type is characterized by its target pathways, receptor distribution, and effects (Table 7 .42). α1-Adrenergic receptors are activated by adrenaline and noradrenaline with about equal potency. Numerous splice variants of α1-adrenoceptors exist. α1d-Adrenoceptors form heterodimers with α1bor α2-adrenoceptors [5] . Signaling is carried out predominantly via Gq/11, but also Gi/o, Gs, G12/13, and Gh subunit. 67 α1-Adrenoceptor subtypes show differences in regulation and coupling efficiency to Ca ++ signaling (α1a > α1b > α1d) and mitogen-activated protein kinase modules α1-Adrenergic receptors of vascular smooth muscle cells as well as those of prostate cause contraction and favor cell growth and proliferation [896] . In heart, Gq-coupled α1-adrenoceptor has positive inotropic effect [897] . Activated Gq excites phospholipase-Cβ that hydrolyzes phosphatidylinositol (4,5)-bisphosphate to 66 . Plasma adiponectin concentration lowers in obesity, insulin resistance, and type-2 diabetes. 67. Subunit Gh consists of the 74-kDa Gα subunit and a 50-kDa β subunit. Gh-proteincoupled α1-adrenoceptor stimulates membrane-bound phospholipase-C [895]. Gs Gs, Gi/o Ad ∼ NAd inositol (1,4,5)-trisphosphate and diacylglycerol. Increased diacylglycerol level stimulates protein kinase-C, but the latter is not significantly involved in cardiac inotropy. Positive inotropy indeed relies mainly on myosin light-chain kinase and RoCK kinase. α1a-Adrenoceptors α1a-Adrenoceptor, or α1c-adrenoceptor, 68 undergoes alternative splicing that generates transcript variants and isoforms with distinct C-termini, but similar ligandbinding properties. α1a-Adrenoceptors do not form heterodimers. α1a-Adrenoceptor mediates G1-S cell cycle arrest that is associated with reduced activities of cyclin-E and cyclin-dependent kinase CDK6 and elevated action of cyclin-dependent kinase inhibitor CKI1b [898] . α1b-Adrenoceptors α1b-Adrenoceptor (encoded by the ADRA1B gene) contributes to the cell cycle progression of many cell types [898] . It can hence acts as a proto-oncogene product. It interacts with subunit-μ2 (AP2M2: adaptor-related protein complex-2 subunit-μ2 or clathrin-associated adaptor protein ClAPM2; but not μ1, μ3, or μ4 subunits of other adaptor complexes) of the clathrin adaptor complex-2 for clathrin-mediated receptor endocytosis [900] . α1b-Adrenoceptor heterodimerizes. 68. A.k.a. AdRa1a, AdRa1c, and AdRa1L. (2) Gi/o and the Gβγ-PI3K-PKB, Gβγ-PLC-PKC, and Gβγ-NOS-PKG pathways; and (3) Gq/11 and the Gβγ-PI3K-PKB pathway (vSMC: vascular smooth muscle cells; bSMC: bronchial smooth muscle cells; CMC: cardiomyocytes; NC: nodal cells; AC: adipocyte; HC: hepatocyte; TC: thrombocyte; PNE: presynaptic nerve ending; A: artery; V: vein; B, C, D, I, and L: bathmo-(nodal excitability [excitation threshold]), chrono-(heart rate), dromo-(nodal conduction velocity), ino-(strength of contraction), and lusitropic (relaxation) effects in cardiac electromechanical coupling; main sources: [736] , Wikipedia). In addition to cardiovascular effects, α1-adrenoceptors also cause smooth muscle contraction of ureter, urethral sphincter, vas deferens, uterus at end of pregnancy, arrector pili muscles, and bronchioles. They induce glycogenolysis and gluconeogenesis in adipose tissue and liver, secretion from sweat glands, and sodium reabsorption from kidney. α2-Adrenoceptors inhibit insulin release and provoke glucagon release in pancreas, and induce contraction of sphincters of the gastrointestinal tract. β1-Adrenoceptors elicit renin release from juxtaglomerular cells and lipolysis in adipose tissue. β2-Adrenoceptors relax urinary sphincter, detrusor urinae muscle of bladder wall, and pregnant uterus, but contract sphincters of the gastrointestinal tract, thicken salivary secretions, induce glycogenolysis and gluconeogenesis, inhibit histamine-release from mastocytes, and increase renin secretion from kidney. α1d-Adrenoceptor (encoded by the ADRA1D gene), like α1a-adrenergic receptor, mediates cell cycle arrest during the G1-S transition [898] . Whereas α1aand α1b-adrenoceptors localize to the plasma membrane, α1d-adrenergic receptors are intracellular. α1b-Adrenoceptor, but not α1a-adrenoceptor, forms heterodimers with α1d-adrenoceptors to cause their translocation to the cell surface [899] . In the developing fetus, cerebral arteries differ from those in adults. Maturational differences in adrenergic-mediated contractility of cerebral arteries result from distinct expression and/or activities of α1-adrenergic receptor subtypes. Once stimulated, they increase the intracellular inositol (1,4,5)-trisphosphate concentration, hence Ca ++ influx, as well as activation of protein kinase-C and extracellular signal-regulated kinases ERK1 and ERK2. In sheep fetuses, α1d-adrenoceptors are expressed at a significantly greater level than α1aand α1b-adrenoceptors [901]. However, α1AR subtype expression is approximately 20% in fetal cerebral arteries compared with that in adults. In fetal cerebral arteries, α1b and α1dAR subtypes contribute to vasocontriction as well as activation of ERK1 and ERK2. α2-Adrenergic receptors are activated by adrenaline and noradrenaline with a potency order (adrenaline > noradrenaline) [5] . α2-Adrenoceptors activate the Gα i/o family of guanine nucleotide-binding regulatory (G) proteins that, in turn, regulates several effectors, such as adenylate cyclase (inhibition), Ca ++ channels (stimulation or inhibition), K + channels and H + -Na + antiporter (stimulation) [902] . α2a-Adrenoceptor mediates the central baroreflex control (hypotension and bradycardia) as well as influence platelet aggregation [5] . α2b Subtype is involved in neurotransmission in the spinal cord. α2c-Adrenoceptor regulates the catecholamine release from adrenal chromaffin cells. α2a-Adrenoceptor is widespread in the body's tissues (brain, vasculature, submandibular gland, and kidney, as well as platelets). Its desensitization after acute stimulation via that phosphorylation by GRKs depends on tissue and species types. It is constitutively palmitoylated. It can interact with α-subunit of eukaryotic initiation factor-2B and 14-3-3ζ protein, as well as spinophilin [902] . α2a-Adrenoceptor is able to form homo-and heterodimer with α2c subtype as well as β1-adrenoceptor. Gi-Coupled α2a-adrenergic receptor (encoded by the ADRA2A gene) impedes insulin secretion, as it causes a reduced exocytosis of pancreatic β cells and insulingranule docking at the plasma membrane [903] . α2b-Adrenoceptor has a very limited expression in humans. It can promote mitogen-activated protein kinase activation. Desensitization relies on phosphorylation by GRK2 or GRK3 and, then, interaction with β-arrestin. On the other hand, scaffold spinophilin (or neurabin-2) tethers the receptor at the cell surface of dendritic cells. In addition, it can form complexes with α subunit of eukaryotic initiation factor-2B and 14-3-3ζ protein [902] . α2c-Adrenoceptor is mainly expressed in the brain. It forms homo-or heterodimer with α2a subtype that can impair GRK phosphorylation and β-arrestin recruitment. It can also interact with β2-adrenergic receptors. In addition, it can build complexes with α subunit of eukaryotic initiation factor-2B and 14-3-3ζ protein [902]. Three βAR subtypes (β1AR-β3AR) can bear distinct regulation and triggers different signaling cascades. Specificity of βARs for G-protein coupling is particularly dictated by the third intracellular loop and C-terminal tail. Intracellular domains of βARs are important for binding of βAR regulatory proteins such as G-proteincoupled receptor kinases and β-arrestins. 69 β-Adrenoceptors possess a set of contacts between helices 3 and 6, the socalled ionic lock that forms a molecular switch for receptor activation. Molecular dynamics simulations of β2-adrenergic receptor show that the ionic lock forms reproducibly [904] . Inactive β2AR conformations vary from formed lock to broken lock, the latter conformation being observed in crystal structures. β1-Adrenergic receptors activate adenylate cyclase and strongly elevate the cAMP concentration, whereas β2 and β3-adrenergic receptors can couple to Gi. They also stimulate many other signaling pathways, particularly mitogen-activated protein kinases. In rat ventriculomyocytes, β2-adrenoceptors couple to Gs (preferentially) and Gi protein, activating either Gs or both Gs and Gi protein [905] . Adenylate cyclase activation by Gi2-coupled β2-adrenoceptors can be done by preventing Gi2 to interact with the inhibitory Gα i site of adenylate cyclase [906] . The control of cAMP transients via degradation by phosphodiesterases (Vol. 4 -Chap. 10. Signaling Pathways) corresponds to a specific procedure for GPCR-cAMP signaling [907] (Table 7 .43). Liganded β2AR recruits a preformed β-arrestin-PDE4d5 complex. Inactivated β1AR forms a signaling complex with another cAMPspecific PDE, PDE4d8, in cardiomyocytes. Ligand-induced dissociation of PDE4d from the β1AR-PDE4d8 complex produces a localized cAMP transient. Gs-Coupled β-adrenergic receptors activate adenylate cyclase. β1-Adrenoceptor is the predominant subtype in heart. It is also a major mediator of lipolysis in adipocytes and renin release from kidney cells. Lastly, it is the principle subtype in certain brain regions, such as cerebral cortex and hippocampus, where it modulates long-term potentiation and, thereby, synaptic remodeling and memory [908] . 69 . Upon agonist stimulation, GRKs are recruited to the plasma membrane and phosphorylate activated βARs. β-Arrestins then bind to phosphorylated βARs to prime a rapid desensitization and receptor internalization. (transducer, scaffold, and effector) and production between β-adrenoceptors (Sources: [907] ). Different modes of interaction with phosphodiesterases (PDE) generate distinct cAMP signals for β-adrenoceptor signaling specificity. Signaling complex PDE4d8 β-Arrestin-PDE4d5 Agonist binding Complex dissociation Complex formation Human β1AR is glycosylated (Asn15). It heterodimerizes with α2aAR that has a lower potency for agonist stimulation of cAMP production. It interacts with Gs, but, unlike β2AR, not with Gi. Its desensitization results from phosphorylation by protein kinase-A and -C. Its phosphorylation by GRK is much less than that of β2AR; hence arrestin binding and desensitization is weaker [908] . Many class-1 PDZ domain-containing proteins link to β1AR, influencing either β1AR endocytosis or signaling. β1-Adrenoceptor indeed connects to Disc large homologs DLg1 70 and DLg4, 71 members of the membrane-associated guanylate kinase (MAGuK) family, as well as MAGuK-like proteins with an inverted domain structure (WW and PDZ domain-containing proteins) MAGI2 and MAGI3 [909] . Protein DLg4 inhibits and MAGI2 stimulates β1AR endocytosis, respectively. The association of MAGI3 with β1AR impairs activation of ERK1 and ERK2 without affecting cAMP generation or internalization. Cystic fibrosis transmembrane conductance regulator-associated ligand (CAL) 72 also connects to the β1AR C-terminus. β1-Adrenoceptor also interacts with GAIP-interacting protein C-terminus product GIPC and RapGEF2. The former regulates β1AR-mediated activation of ERK. The latter promotes the activation of Ras by β1AR. Appropriate β1AR transfer after agonist-promoted endocytosis is needed for resensitization of its signaling pathway. Efficient β1AR recycling requires binding of protein kinase-A-anchoring protein AKAP79 to β1AR C-terminus. Moreover, AKAP79 forms a complex with DLg1 and PKA, the so-called β1AR receptosome [910] . This scaffold allows β1AR recycling and PKA-mediated β1AR phosphorylation (Ser312). In addition, endophilins enhance β1AR internalization [911] . 73 They are involved in clathrin-mediated endocytosis. Endophilins colocalize with endocytic reg- Endophilins are also identified as Src homology domain-3-containing proteins (SH3P) and SH3-domain GRB2-like proteins (SH3GL). Endophilin-1 (a.k.a. Een, SH3GL1, and SH3P8), -2 (a.k.a. EenB1, SH3GL2, and SH3P4), and -3 (a.k.a. EenB2, SH3GL3, and SH3P13) bind to β1-adrenoceptors. ulators, such as GTPase dynamin, phosphatase synaptojanin, and amphiphysin, an adaptor of the cytoplasmic surface of synaptic vesicles in nerve terminals. β2-Adrenoceptor β2-Adrenergic receptors are synthesized in most cell types. However, β2-adrenoceptor is the predominant type in the lung, smooth muscle, and liver. In lungs, β2AR resides on airway smooth muscle and epithelial cells. It is also detected in cardiomyocytes and inflammatory cells. It targets MAPK and PI3K kinases. In cardiomyocytes, β2AR-mediated activation of these kinases can counter apoptosis caused by chronic β1AR stimulation [912] . Phosphorylation by GRK followed by β-arrestin binding leads to β2AR endocytosis. MicroRNA Let7f represses the translation of the ADRB2 mRNA, thereby attenuating the production of β2-adrenergic receptor under stable exposure in a given time range of agonists [913] . 74 On the other hand, during agonist activation as well as long-term, chronic agonist exposure, the Let7f level decays (positive feedback loop that relieves the Let7f-mediated repression of ADRB2 translation). β2-Adrenergic receptor is activated by the hormone and neurotransmitter adrenaline that is strongly involved in cardiovascular and pulmonary physiology. Ligandbinding site is accessible by conformational changes in the extracellular loop [914] . The β2AR basal (activator-independent) activity (and structural instability) involves interactions (hydrogen bonds and charge interactions, the so-called ionic lock) between cytoplasmic ends of transmembrane helices TM3 and TM6 [915] . β2-Adrenergic receptor phosphorylation by G-protein-coupled receptor kinases and protein kinase-A rapidly desensitizes these receptors, hence attenuating airway smooth muscle cell relaxation and bronchodilation. G-protein-coupled receptor kinases GRK2 and GRK5 with subsequent binding of β-arrestins reduce liganded β2AR-Gs coupling. Protein kinase-A also lowers β2AR-Gs coupling, but promotes β2AR-Gi coupling [916] . Protein kinase-A primes the major desensitization pathway in airway smooth muscle cells, especially at low agonist concentrations. β2-Adrenergic receptor interacts with the Na + -H + -exchanger regulatory factors NHERF1 (or SLC9a3r1) and NHERF2 (or SLC9a3r2) to regulate Na + -H + exchanger as well as β2AR postendocytic sorting following agonist-promoted internalization [909] . The recruitment of NHERF1 to β2AR precludes inhibition by NHERF1 on Na + -H + exchanger NHE3 (or SLC9a3) in renal cells. Interactions between NHERF1 with β2AR are regulated by agonists. Endocytosis of β2-adrenoceptors is regulated by agonist concentration and mediated by pre-existing clathrin-coated pits. β2-Adrenoceptors regulate the duration 74. The so-called "agonist-promoted downregulation" corresponds to the loss of the receptor response under prolonged exposure to an endo-or exogenous agonist. The miR-mRNA interaction prevents mRNA translation into a protein. The Let7f microRNA thus contributes to set the baseline of expression of β2-adrenergic receptors. The 3 untranslated region of ADRB1 and ADRB2 mRNAs does not contain Let7f-binding sites for all subtypes (βARspecific mechanism) [913] . Two other microRNAs, miR15 and miR30, have predicted binding sites in the 3 UTR of ADRB2 mRNAs. between clathrin coat deposition and membrane scission by a mechanism that involves receptor scaffolding to cortical actin [11] . Activation of protein kinase-A by β2ARs and subsequent receptor phosphorylation regulates Rab4-dependent, rapid recycling, but not slower recycling. In rodent cardiomyocytes, Gs-coupled β2AR are confined to transverse tubules, whereas β1ARs are distributed across the entire cell surface. Both respond to the same ligands and couple to the same effectors, but they elicit different cellular responses. 75 In cardiomyocytes of a rat model of chronic heart failure, β2ARs redistribute from the transverse tubules (sites of coupling between the plasma membrane depolarization to calcium entry) to the plasma membrane [917] . A diffuse receptor-mediated cAMP signaling then results in contrast to a spatially confined β2AR-induced cAMP signals in normal condition. Loss of proper PKA localization is also observed in human heart failure. The classical receptor theory postulates that receptors switch between inactivation and activation. Full agonists stimulate all of the signaling pathways of a given receptor to the same extent. The concept of "functional selectivity", or "biased signaling", states that a natural ligand can have distinct performance on different triggered signaling pathways. The endogenous ligands of β2-adrenergic receptor are adrenaline and noradrenaline. β2-Adrenergic receptor has 2 primary modes of activation of adenylate cyclase either via Gs subunit or, upon endocytosis, via β-arrestins and mitogen-activated protein kinases. The type of conformational change differs according to the type of agonists [918] . Both ligand types activate with an almost similar efficiency Gs and adenylate cyclase. However, noradrenaline is less efficient in signaling to β-arrestin. Therefore, noradrenaline is more tightly coupled to the Gs-ACase-cAMP pathway than signaling initiated during receptor endocytosis. In humans, whereas β1AR and β2AR are ubiquitous, β3AR is essentially produced in the digestive tract and adipose tissue. In humans, β3AR abounds in brown, but not white adipocytes. β3-Adrenergic receptor regulates metabolic and endocrine functions, using the Gs-cAMP-PKA and PKC-P38MAPK pathways [919]. Sustained action of insulin reduces the density of β3AR, the major βAR subtype of adipocyte, without effect on β1AR (β2AR is undetectable) [920] . Activated β3AR stimulates lipolysis and thermogenesis in human adipocytes. 75 . Upon stimulation, β1ARs generate positive ino-and chronotropic effects via phosphorylation by PKA of regulators of the intracellular concentration of calcium. β1-Adrenoceptors produce cytosolic cAMP that diffuses toward sarcomeres to enhance contractility and frequency. In heart failure, β1AR causes cell apoptosis. β2-Adrenoceptors are also coupled to Gs subunit and adenylate cyclase, but not calcium regulators and myofilaments that control contraction. In addition, β2AR activity opposes β1AR signaling because they switch sequentially from Gs to Gi, hence producing different cAMP pools in the cardiomyocyte and subcellular compartmentation of signals. Table 7 .44. Distinct action of β1and β2-adrenergic receptors in cardiac physiology and pathology (Sources: [922, 923] ; ACase: adenylate cyclase; cAMP: cyclic adenosine monophosphate; PI3K: phosphoinositide 3-kinase; PKA: protein kinase-A). β1and β2-adrenoceptors are the predominant subtypes expressed in hearts. Both β1and β2adrenoceptors trigger the Gs-ACase-cAMP-PKA pathway. β2-Adrenoceptor also activates Gi protein that prime the Gα i Gβγ-PI3K-PKB pathway. The latter competes with the former for protein phosphorylation (calcium channels Ca V 1, phospholamban, and troponin) via PI3K during acute stimulation: β2AR-Gi coupling partially inhibits β2AR-Gs complex activity, i.e., positive inotropy (I+) and lusitropy (L+). Persistent β1AR stimulation induces cardiomyocyte hypertrophy and apoptosis via protein kinase-A-independent activation of calmodulindependent protein kinase-2 (CamK2). On the other hand, persistent β2AR activation exerts a survival effect in cardiomyocytes via protein kinase-B (PKB). Gs-ACase-cAMP-PKA pathway Gs-ACase-cAMP-PKA pathway The sympathetic nervous system stimulates blood circulation in response to stress or exercise by activating β1and β2-adrenoceptors, which activate G-protein subunit stimulatory for adenylate cyclase. Yet, a sustained β-adrenoceptor stimulation can yield pathological cardiac remodeling. Cardiac β1-adrenoceptors are coupled to Gs and β2-adrenoceptors to both Gs and Gi (Table 7 .44). The duration and magnitude of receptor expression determine the cardiomyocyte fate. β2-Adrenoceptors, like most GPCRs, exist at very low concentrations in the sarcolemma. In heart, α1aand α1b-adrenoceptors have distinct roles [924] . Overexpression of α1bARs depresses heart contractile response to βAR activation via Gi and predisposes heart to hypertrophy, whereas α1aAR overexpression increases cardiac contractility and improves outcomes after pressure overload or myocardial infarction. Both subtypes hinder the generation of inositol trisphosphate and protect from arrhythmogenesis in early postischemic reperfusion. In cardiomyocytes, β1and β2-adrenergic receptors activate mitogen-activated protein kinases P38MAPK and ERK, respectively. α1-Adrenergic receptors can activate the PLC-PKC pathway, stress-activated protein kinases (P38MAPK and Jun N-terminal kinase), Rho and Rac GTPases, and Ca ++ -calmodulin-dependent protein kinase [925] . A single or several (1-7) ryanodine receptors of a given couplon release Ca ++ sparks from the sarcoplasmic reticulum in response to its natural trigger, the Ca ++ sparklet generated by a single Ca V 1 channel. β-Adrenergic receptors regulate the pace and strength of heart contraction, as they cause both prolonged Ca V 1 channel opening duration and accelerated channel coupling. They raise the coordination between Ca V 1 channels and ryanodine receptors via the PKA pathway. The enhanced synchronization of these 2 types of Ca ++ channels generates higher and quicker Ca ++ transients to support stronger and faster heart contraction. Channel synchronization relies on: (1) synchronized Ca V 1 channel activity (trigger); (2) synchronized initiation of ryanodine receptor activity (accelerated activation kinetics); and (3) synchronized ryanodine receptor activity (recruitment) [926] . Protein kinase-A phosphorylates Ca V 1 channels, thereby reducing their activation latency and enhancing Ca V 1-mediated Ca ++ influx that, in turn, by improving connection between the 2 channel types elicits greater Ca ++ release from the sarcoplasmic reticulum via ryanodine receptors, which are also phosphorylated. Because the Ca ++ conductance of ryanodine receptors is not sensitive to phosphorylation, β-adrenoceptors increase the kinetics and number of ryanodine receptors that respond nearly simultaneously to the Ca ++ flux delivered by Ca V 1 channels. Acute β1-adrenoceptor stimulation triggers the cAMP-PKA pathway and modulates the cardiac excitation-contraction coupling [922] . On the other hand, sustained β1-adrenoceptor stimulation elicits cardiomyocyte hypertrophy and apoptosis via increased intracellular calcium concentration and Ca ++ -CamK2 activity independently of PKA. However, a persistent β2AR activation can exert a survival effect in cardiomyocytes via protein kinase-B. Activation of ERK1, ERK2, and P38MAPK by the Gi subunit also favors β2AR-mediated anti-apoptotic effect in adult rat cardiomyocytes. Several transcriptional targets of α1-adrenergic receptors have been identified in cardiomyocytes, particularly α-actin, β-myosin heavy chain, atrial natriuretic factor. 76 Four adrenoceptor types are involved in the regulation by catecholamines of lipolysis in adipocytes. The control of adenylate cyclase activity involves stimulatory β1-, β2-, and β3-adrenoceptors. On the other hand, α2-adrenoceptors reduce intracellular cAMP content, thereby reducing lipolysis. Adipocytes, like platelets, possess α2arather than α2d-adrenoceptors, at least in rabbits [928] . In humans, 76. α1ARs target transcription factors, such as Jun (Jun stands for avian sarcoma virus-17 oncogene; Japanese: 10 [Ju] + 7 [Nana]), Fos (cellular Finkel Biskis Jinkins murine osteosarcoma virus sarcoma oncogene), and EGR1 (early growth response factor-1), as well as transcriptional corepressor CARP. The zinc finger-only protein Zfp260 of the Krüppel family of transcriptional regulators, highly expressed in the embryonic heart, is downregulated during postnatal development. Protein Zfp260 is a nuclear effector of α1-adrenergic receptors. It is also a transcriptional activator of atrial natriuretic factor and a cofactor for cardiac regulator GATA4, used in MAPK signaling [927] . adrenaline stimulates and inhibits lipolysis in visceral (omental) and subcutaneous adipocytes that have a high and low βto α2-adrenoceptor ratio, respectively. In white adipose tissues, types of adrenoceptors on the cell surface depends on mammal species. Adipocytes produce mainly α2and few β3-adrenoceptors in humans, and conversely in rodent adipocytes [929] . Adrenaline controls lipid mobilization, as it activates antilipolytic α2ARs in human subcutaneous adipocytes during exercise [930] . Estrogens promote and maintain the female type of adipose tissue distribution. They attenuate the lipolytic response by increasing the number of antilipolytic α2a-adrenoceptors only in subcutaneous fat depots, thereby shifting the assimilation of lipids from omental depots to subcutaneous adipose tissues [931] . In addition, increased [Ca ++ ] i inhibits lipolysis induced by β-adrenoceptor activation via phosphodiesterase inhibition of cAMP signaling. In adipocytes, insulin attenuates β1-adrenoceptor-mediated lipolysis via activation of protein kinase-C. The PKCβ1 isoform interacts with β1AR signaling to decay lipolysis [932] . β3-Adrenoceptors are major mediators of the lipolytic and thermogenic effects of catecholamines. β3-Adrenoceptor activates extracellular signal-regulated kinases ERK1 and ERK2 via epidermal growth factor receptor and Src kinase (Vol. 4 -Chap. 3. Cytosolic Protein Tyr Kinases). This pathway in association with protein kinase-A contributes to maximal β3AR-stimulated lipolysis. Vimentin of intermediate filaments interacts with the β3AR-ERK pathway, whereas actin microfilaments and microtubules do not affect lipolysis and ERK activation [933] . Hepatocytes express adrenergic receptors that modulate liver regeneration, hepatocyte proliferation, glycogenolysis, gluconeogenesis, urea synthesis, and fatty acid metabolism. Under basal conditions, the liver supplies glucose only via gluconeogenesis to spare glycogen stores, whereas in situations of enhanced demands primed by glucagon or sympathetic signal, additional glucose release occurs via glycogenolysis. Glucagon is the main factor that increases cAMP level, which is correlated with the rate of glycogenolysis and exercise intensity [934] . Both αand β-adrenergic receptors exist in liver. However, α-adrenergic stimulation is much more important than that of β-adrenoceptor to prime formation of cAMP via adenylate cyclase for glycogenolysis, gluconeogenesis, and ureogenesis. Adrenergic control of the hepatic function is mainly due to α1-adrenoceptors. Transactivation from α1AR of epidermal growth factor receptor involves Gq/11, Src kinase, signal transducer and activator of transcription STAT3, and the Src-STAT3 complex (pathway crosstalk) [935] . Normal and regenerating hepatocytes express α1b-, but not α1a-adrenoceptors [936] . Liver regeneration after partial hepatectomy involves β-adrenoceptors in the initial stage and afterward α1-adrenoceptors [937]. G-protein-coupled receptors can signal without G proteins. Stimulated β2-adrenoceptor recruits β-arrestin that forms a complex with phosphodiesterase PDE4 to regulate plasmalemmal PKA activity, which phosphorylates β2-adrenoceptor [938]. Arrestin-mediated uncoupling of G-protein-mediated signaling hampers Gs activation and switches to Gi stimulation that activates extracellular signal-regulated kinase ERK2. Angiotensins (ATn1-ATn4) act mainly via angiotensin type-1 (AT 1 ) and type-2 (AT 2 ) receptors (Table 7 .45). Endogenous ligands are mostly angiotensin-2 and -3, whereas angiotensin-1 is weakly active in some systems [5] . In fact, different types of angiotensin receptors exist: AT 1A , AT 1B , and AT 2 to AT 4 . The AT 4 receptor is activated by angiotensin-4, an angiotensin-2 metabolite. This specific receptor of angiotensin-4 resides in the brain and kidney. Specific receptors for angiotensin-2 in adrenal zona glomerulosa, 77 vascular smooth muscle, kidney, brain, and anterior pituitary gland 78 exhibit generally similar binding properties. Angiotensin receptors of the anterior pituitary are not affected by changes in sodium balance in opposition to adrenal and vascular receptors [939] . In the brain, angiotensin receptors localize to several regions, although they are particularly concentrated in the circumventricular organs. 79 The angiotensin receptors of the renal cortex are localized in glomeruli, whereas in the renal medulla they are distributed diffusely. Initiated signaling pathways include, in particular, receptor Tyr kinases (e.g., PDGFR and EGFR), IRS1, and cytosolic Tyr kinases, such as Src, Janus, and focal adhesion kinases, as well as mitogen-activated protein kinase. 77. Zona glomerulosa is the superficial layer of the adrenal cortex. 78. Among endocrine cell types -corticotrophic, gonadotrophic, lactotrophic, somatotrophic, and thyrotrophic cells -of anterior pituitary gland, angiotensin-2 receptors are located in corticotrophs and lactotrophs. 79. Circumventricular organs surround the brain ventricular network, primarily the third and fourth ventricles. They correspond to the few cerebral sites that have an incomplete bloodbrain barrier, so that neurons can sense various blood compounds, particularly hormones. These organs secrete different neurotransmitters, hormones, and cytokines. Circumventricular organs include pineal gland (circadian rhythm), subfornical organ and organum vasculosum of the lamina terminalis (body fluid regulation), choroid plexi, area postrema (toxin detection), median eminence (regulation of anterior pituitary activity), subcommissural organ (somatostatin secretion), and posterior pituitary (oxytocin and vasopressin sensing). 7.13.6.1 AT 1 Type-1 receptor AT 1 mediates vasoconstriction, secretion of aldosterone and vasopressin, cardiac contractility and hypertrophy, augmented peripheral noradrenergic activity, vascular smooth muscle cells proliferation, decreased renal blood flow, renin inhibition and sodium reuptake in kidneys, modulation of central sympathetic nervous system activity, central osmocontrol, and extracellular matrix formation. Angiotensin-2 receptor AT 1 activates: (1) Gα q/11 , Gα i/o , and Gα 12/13 subunits of guanine nucleotide-binding (G) proteins; (2) Angiotensin-2 receptor AT 1 predominantly and robustly couples to Gq/11 subunit family. Once bound to AT 1 receptor, angiotensin-2 stimulates vascular cells. In vascular smooth muscle cells, upon AT 1 binding, angiotensin-2 activates phospholipases as well as kinases and various oxidases. G12-Coupled AT 1 can stimulate monomeric GTPase Rho and inhibit PDGF-activated Rac GTPase. The Gβγ subunit associated with G12 and/or Gq/11 family subunits mediates PLC activation by angiotensin-2. Associated with G12 subunit, it also intervenes in angiotensin-2-induced PLD1 activation, possibly via Src-and RhoA-dependent mechanisms. 82 In cultured rat vascular smooth muscle cells, angiotensin-2-mediated activation of phospholipase-D1 via Gβγ provides a major source of sustained generation of second messengers. The G12 subunit contributes to PLD activation, whereas Gi and Gq/11 have no effect [941] . Small GTPase RhoA participates in this signaling pathways. Hydrolysis of phosphatidylcholine by PLD causes a strong production of phosphatidic acid and subsequent generation of diacylglycerol by phosphatidic acid phosphatase. Diacylglycerol contributes to continuous PKC activation. Phosphatidic acid contributes to the activation of NADH/NADPH oxidase that mediates the hypertrophic effects of angiotensin-2. Angiotensin-2 promotes the association of scaffold proteins (paxillin and talin), leading to focal adhesion. It also stimulates NADPH oxidase via AT 1 receptor. Resulting reactive oxygen species are components of ATn2-mediated signal transduc-80. Once bound to agonist, AT 1 can recruit a complex of non-receptor protein tyrosine kinase and phosphatase, i.e., Janus kinase JaK2 and PTPn11 that facilitates JaK2 phosphorylation (activation). This additional G-protein-independent AT 1 -JaK2 pathway complements signaling primed by G-protein subunit Gq that can further potentiate the JaK-STAT signaling via Ca ++ import in the cytosol [835] . 81. Phospholipase-Cβ1, then PLCγ1, are activated by corresponding stimulated G proteins within 5 s [940] . Diacylglycerol and inositol trisphosphate are thus generated, the production ending within minutes. The transient PLC activation is followed by a sustained activation of phospholipase-D via G proteins. Concomitant stimulation of phospholipase-A2 initiates arachidonic acid signaling. 82. The Gβγ subunit can activate PLD by binding to Rho or ARF GTPases. tion in vascular smooth muscle cells. In addition, AT 1 and bradykinin B2 receptors can heterodimerize [5] . The AT 2 receptor is much less abundant than AT 1 in adult tissues. It is upregulated in pathological conditions. 83 Receptor AT 2 counteracts several growth responses initiated by AT 1 . Receptors AT 1 and AT 2 stimulates and inhibits ERK1 and ERK2, respectively. Type-2 receptor AT 2 has a more restricted expression pattern than the AT 1 receptor. It intervenes in cell growth and differentiation and tissue development. It is highly expressed in fetuses, whereas in adults, it is confined to heart, vascular smooth muscle, brain, adrenal cortex, uterus, and ovarian follicles. Activated AT 2 dilates blood vessels, inhibits growth, and causes apoptosis. It provokes a rapid activation of Gi and Go (but not of Gq and Gs) [943]. Angiotensin-2 causes cell proliferation, hypertrophy, vasoconstriction, and fibrosis [944] . On the other hand, angiotensin (1−−7) , an active heptapeptide formed from angiotensin-1 and-2, antagonizes angiotensin-2. It provokes vasodilation, as it causes the release of nitric oxide and vasodilatory prostaglandins, and can amplify bradykinin effect [944] . In addition, it has a potent natriuretic and antifibrotic action. It also inhibits the growth of vascular smooth muscle cells and cardiomyocytes. Angiotensin (1−−7) is preferentially formed from angiotensin-2 by Angiotensinconverting enzyme-2 (ACE2). Angiotensin-2 reduces ACE2 synthesis and activity in cardiac myocytes and fibroblasts. Endothelin-1 also attenuates ACE2 synthesis in cardiomyocytes. Decrease in ACE2 production by angiotensin-2 and endothelin-1 requires mitogen-activated protein kinase kinase MAP2K1 and extracellular signalregulated kinases ERK1 and ERK2. Atrial natriuretic peptide reverses ATn2-and ET1-mediated ACE2 downregulation, when it acts simultaneously with these factors (but not alone). 84 Angiotensin (1−−7) impedes ATn2-and ET1-mediated ACE2 downregulation (but not its synthesis) via a specific AT (1−−7) receptor [945]. 85 Stimulated AT (1−−7) activates the PI3K-PKB signaling cascade and endothelial nitric oxide synthase (NOS3) 83. The AT 1 receptor is involved in the evolution of aortic aneurysm in Marfan syndrome, an autosomal dominant connective tissue disorder characterized by heterozygous mutations in the FBN1 gene that cause a deficiency of fibrillin-1. Transforming growth factor-β signaling augments. Manifestations include aortic aneurysm, emphysema, degeneration of the atrioventricular valves, and myopathy. On the other hand, AT 2 confers protection [942] . 84. Atrial natriuretic peptide, a peptide hormone released mainly from the atrium, activates the guanylate cyclase-A receptor (NPR1 or NPRa; Sect. 6.4.1). Activated NPR1 produces cGMP from GTP. Second messenger cGMP causes natriuresis and hypovolemia, and impedes the proliferation of vascular smooth muscle cells. increases ANP release at high atrial pacing via the PI3K-PKB pathway. Conversely, Na + -H + exchanger-1 and Ca ++ -calmodulin-dependent kinase CamK2 prevent the augmentation of high atrial pacing-induced ANP secretion by ATn (1−−7) . Angiotensin (1−−12) can trigger a vasoconstriction via AT 1 receptor. It can serve as a substrate for the formation of angiotensin-2 and angiotensin (1−−7) , according to the available, local type of angiotensin-converting enzyme. In the brain, its concentration is higher than those of angiotensin-1 and -2. It is produced in the nucleus tractus solitarius (baroreceptor reflex center), but do not affect the autonomic control of the cardiac frequency in normal conditions. However, angiotensin (1−−12) may be active in hypertension [946] . Exogenous angiotensin (1−−12) is converted into angiotensin-2 within the nucleus tractus solitarius. It can cause a transient depressor response [946] . The vasculature is characterized by a low density of AT 1 in endothelial cells (ecAT 1 ) and its relative abundance in vascular smooth muscle cells (smcAT 1 ). Endothelial cell activation by angiotensin-2 via ecAT 1 reduces the tone of vascular smooth muscle cells, as it elevates expression of endothelial nitric oxide synthase [947] . Therefore, angiotensin-2 stimulation of ecAT 1 reduces smcAT 1mediated vasoconstriction. The AT 1 receptor contributes to vascular relaxation in cerebral resistance arteries via transactivation of epidermal growth factor receptor kinase and extracellular signal-regulated protein kinases ERK1 and ERK2 [948] . In hearts, pathways leading to ERK activation differ according to cell types. In cardiac fibroblasts, angiotensin-2 activates ERKs via Gβγ subunit of Gi, Src kinase, adaptors SHC and GRB2, and Ras GTPase. In cardiomyocytes, mechanical stretch-induced ERK activation that can occur even in the absence of angiotensin-2 involves Gq and protein kinase-C [949]. Cardiomyocyte stretch indeed leads to conformational change and activation of AT 1 independently of angiotensin-2 [950, 951]. The AT 1 receptor is able to detect mechanical stress and transduces it into biochemical signals. Mechanical stretch induces association of AT 1 receptor with Janus kinase-2 and translocation of G proteins into the cytosol. Angiotensin-2 operates as a potent hypertrophic agent for cardiomyocytes that promotes production of TGFβ1 and atrial natriuretic factor [952] . 86 The AT 2 receptor is involved in maladaptive cardiac hypertrophy. On the other hand, cardiac 86. Angiotensin-2 can, hence, indirectly cause cell division, fibrosis, cytokine infiltration, αto β-myosin heavy chain switch, and stimulation of the renin-angiotensin system. Interactions among cardiomyocytes, fibroblasts, and vascular cells are sources of secreted signals. Locally produced factors, such as growth factors FGF2, IGF1, TNFα, cardiotrophin-1, and endothelin, can also trigger cardiomyocyte hypertrophy. In addition, MAP3K7 can provoke heart failure. AT 1A receptor causes cardiac fibrosis, but not hypertrophy [953] . Transforming growth factor-β1 isoform couples work load and angiotensin-2 to cardiac adaptative hypertrophy Angiotensin-2 produces reactive oxygen species in vascular smooth muscle cells and cardiomyocytes. In rat neonatal cardiomyocytes, angiotensin-2 binds to Gα 12/13coupled AT 1 that primes a cascade with the following mediators, which operate successively: (1) small GTPase Rho and RoCK kinase; (2) small GTPase Rac; (3) NADPH oxidase; (4) reactive oxygen species; and (5) JNK and P38MAPK [954]. Angiotensin-2 acts as a pro-inflammatory agent and growth factor in the liver. After injury to the liver, it assists in tissue repair by stimulating: (1) hepatocytes and hepatic stellate cells to synthesize extracellular matrix proteins and secrete cytokines and (2) myofibroblasts to proliferate. Angiotensin-2 activates nuclear factor NFκB via AT 1 and caspase recruitment domain-containing proteins CaRD10 and B-cell lymphoma/leukemia protein BCL10 as well as paracaspase mucosa-associated lymphoid tissue lymphoma translocation protein MALT1 [955]. 87 Apelin APJ receptor 88 (Sect. 6.9.2) responds to apelins [741], mainly long apelin 36 and short apelin 13 89 (potency order apelin 13 > apelin 36 [5] ). Main transducers are subunits of the Gα i/o family of guanine nucleotide-binding (G) proteins. The hydrophobic region of APJ is similar to that of the angiotensin receptor [956] . The APJ receptor is particularly produced in the cardiovascular and central nervous systems, and in the latter by neurons, oligodendrocytes, and astrocytes [958] . 90 Apelin is also observed in adipocytes as well as gastric mucosa and Kupffer cells in the liver [961]. 87. Caspase recruitment domain-containing proteins (CaRD9 to -11 and -14, as well as BCL10) operate as upstream regulators in NFκB signaling. Proteins CaRDs interact with BCL10 to act as NFκB signaling complexes. Protein BCL10 forms a complex with MALT1 to synergize NFκB activation. 88. The APJ receptor is encoded by chromosome 11. 89. Apelin derives from a precursor, preproapelin (77 amino acids). Apelins include long types (e.g., 36 amino acids; apelin (42−−77) ) expressed in lungs, testes, and uterus, and short types (e.g., 13 amino acids; apelin (65−−77) ) produced in mammary glands, which also synthesize the long apelin type [957] . Long and short forms of apelin differently interact with APJ receptor. 90. Apelin serves a neuropeptide in the hypothalamus, especially in the supraoptic, paraventricular, and arcuate nuclei. Apelin-containing cell bodies of the supraoptic and paraventricular nuclei, such as arginine vasopressin-and oxytocin-containing magnocellular neurons, project toward the internal layer of the median eminence and posterior pituitary (neurohypophysis). In the central nervous system, the apelin-APJ complex helps maintain the fluid homeostasis and regulates vasopressin release from the hypothalamus [959] . In response to osmotic stimulation, the release of arginine vasopressin, leucine enkephalin ( Leu enkephalin), and dynorphin from neurons of the supraoptic and paraventricular nuclei is augmented. 91 Apelin targets vasopressinergic neurons and inhibits their activity, thus impeding arginine vasopressin release. Apelin and arginine vasopressin that coexist in magnocellular neurons have opposite effects (autocrine somatodendritic feedback loop) to maintain body fluid homeostasis. In the cardiovascular system, apelin is primarily expressed in vascular and endocardial endothelial cells (at least in recesses of the right atrium) [962] . 92 Cardiomyocytes can also express apelin. Apelin receptors on vascular smooth muscle and endothelial cells as well as cardiomyocytes are activated by the family of apelin peptides [963] . They operate in both auto-and paracrine signalings. Apelin, a direct vasoconstrictor, 93 can mediate vasodilation via nitric oxide [961] . In fact, the endothelium-dependent vasodilation results from a prostanoid-dependent mechanism [963] . In mammary artery, the 3 main forms of apelin -N-terminal pyroglutamylate pGlu apelin 13 , apelin 13 , and apelin 36 -cause vasodilation, when the endothelium is functional. These regulators act with comparable potency in a concentration-dependent manner. In human hearts, apelin peptides are among the most potent endogenous positive inotropic agents [963] . The N-terminal pyroglutaminated apelin 13 is the predominant isoform in hearts from patients with coronary artery disease. Apelin acts on neighboring cardiomyocytes and exerts positive inotropic effect via Na + -H + exchanger NHE1 (or SLC9a1), Na + -Ca ++ exchanger (reverse mode), phospholipase-C, and protein kinase-C [739]. 94 Autocrine signaling by apelin and APJ receptor in mouse left ventriculomyocytes plays a modest role in basal conditions, but becomes significant under stress, as they both improve sarcomeric shortening and velocity of contraction and relaxation without change in calcium transient [964] . Apelin is also involved in angiogenesis, as it enlarges developing blood vessels through which the flow rate is higher upon stimulation by the Ang1-TIE2 complex [965] . 95 Angiopoietin-1 synthesized by mural cells and migrated hematopoietic stem cells activates its receptor TIE2 to generate apelin production in sprout endothelial cells. 91 . Hypothalamic magnocellular neurons of the paraventricular and supraoptic nuclei synthesize not only vasopressin and oxytocin, but also tyrosine hydroxylase (enzyme of catecholamine synthesis), as well as galanin, dynorphin, and cholecystokinin. High plasma osmolality induces increased mRNA levels for vasopressin, oxytocin, Tyr hydroxylase, galanin, dynorphin, and cholecystokinin [960] . 92. Apelin is produced by endothelial cells of, at least, large blood vessels and vessels of heart, kidneys, adrenal glands, and lungs. 93. Apelins are potent vasoconstrictors in endothelium-denuded saphenous veins and mammary arteries. 94. Apelin is downregulated in chronic ventricular pressure overload. 95. Apelin binds to APJ to enlarge blood vessels. Expression of APJ is enhanced by VEGF. The APJ receptor is coupled to Gα i1 or Gα i2 , but not Gα i3 [966] . Apelin inhibits adenylate cyclase and increases the phosphorylation of ERK or PKB. Apelin efficiency in inhibiting adenylate cyclase depends on: (1) activation and deactivation kinetics of the apelin type and (2) cell type. Activation of apelin receptors by short fragments is transient as APJ is quickly internalized. The duration of desensitization depends on the rate of receptor recycling to the plasmalemma (∼1 h). Long subtypes produces a sustained activation. Bile acid receptor GPBAR1 96 responds to bile acids produced in the liver with the following potency order [5]: lithocholic acid > deoxycholic acid > chenodeoxycholic acid, cholic acid. It activates deiodinases that convert prohormone thyroxine (T4) into active hormone tri-iodothyronine (T3). It signals via Gs and cAMP as well as mitogen-activated protein kinase. Bombesin receptors (BB 1 -BB 3 ) are activated by bombesin homologs 97 gastrinreleasing peptide (GRP) and neuromedin-B and -C [5]. 98 These receptors couple primarily to Gq/11 subunit family of G proteins. Activated BB 1 and BB 2 receptors stimulate tissue growth, smooth muscle cell contraction, and secretion. The BB 3 receptor may intervene in energy balance and control of body weight [5]. Bradykinin receptors (B 1 -B 2 or B1R-B2R) are activated by the nonapeptide bradykinin and its derived peptides kinin and kallidin (Table 7 .46). 99 Kinins are polypeptides formed locally after tissue damage and inflammation. They provoke smooth muscle contraction as well as vasodilation via the release of nitric oxide and prostaglandins by endothelial cells, increase the vascular permeability, and exert a mitogenic effect using the mitogen-activated protein kinase modules. Kinins and their derivatives generated from kininogens by kallikreins and other serine peptidases compose the kallikrein-kinin system. Kallidin is a bioactive kinin that possesses an additional lysine residue at the N-terminal end, hence its name Lys bradykinin. This substrate of carboxypeptidase-M and -N signals through the kinin receptor. It is formed in response to injury from kininogen precursors by 96. A.k.a. M-BAR, GPCR19, GPR131, BG37, and TGR5. 97. Bombesin is a tetradecapeptide originally isolated from skins of amphibians. 98. A.k.a. GRP 18−−27 . 99. The B 1 receptor is expressed at a much lower level than B 2 in mice kidneys, but it is induced upon stimulation by endotoxins or cytokines. kallikreins. Kallidin can be converted to bradykinin by aminopeptidase. Bradykinin and kallidin are produced by the proteolysis of high-or low-molecular-weight kininogen by plasma and tissue kallikrein, respectively. Bradykinin binds and stimulates at least the 2 bradykinin receptors: B 1 and B 2 . The B 2 receptor is constitutively expressed in various cell types. It relays the majority of the vascular actions of bradykinin and kallidin. On the other hand, the B 1 receptor is scarce in normal tissue and expressed during tissue damage (e.g., inflammation and ischemia), by exposure to pro-inflammatory cytokines, growth factors, or oxidative stress, in various cell types, such as vascular smooth muscle and endothelial cells, cardiomyocytes, fibroblasts, and neurons. Bradykinin receptors are coupled to Gq and/or Gi subunits depending on the cell type. Carboxypeptidases remove the C-terminal arginine of bradykinin and kallidin to generate desArg9BK (or BK (1−8) ) and desArg10kallidin (also denoted as Lys BK (1−8) or Lys desArg9BK), respectively. Receptor B 1 is preferentially activated by the metabolites BK (1−8) and Lys BK (1−8) peptides. Angiotensin-2 and endothelin-1 increase the production of B 1 receptor in cardiovascular diseases. These pro-oxidative peptides cooperate via ROS, PI3K, and NFκB on rat vascular smooth muscle cells using AT 1 and ET A receptors [967] . Newly synthesized B 1 receptors can activate the MAPK module. Once the AT 1 receptor is activated, the effect of angiotensin-2 can be mediated by the subsequent ET1 release and ET A stimulation. Regulator of G-protein signaling RGS4 can serve as GTPase-activating proteins for both Gi and Gq (but not for Gs and G12). The RGS4 protein attenuates Gi-mediated inhibition of cAMP synthesis and impedes Gq-mediated activation of phospholipase-Cβ [969] . Both Gq and Gi subunits are able to activate phospholipase-C, the latter via Gβγ. Bradykinin GPCRs can signal without G proteins. In renal cells, the B 2 receptor interacts with protein Tyr phosphatase PTPn11 to prevent cell proliferation [970]. Angiotensin-1-converting enzyme (ACE) is a dipeptidyl carboxypeptidase that removes 2 amino acids from the C-terminus of inactive angiotensin-1 to convert it into active angiotensin-2. Moreover, ACE is also a kininase that converts active vasodilatory kinins into inactive metabolites, as it also deletes 2 amino acids from their C-termini. In humans, the polymorphism of the Ace gene due to the presence or absence of an Alu retrotransposon in intron 16 is associated with up to a 2-fold difference in relative plasma ACE concentration. 100 The kallikrein-kinin system that partly mediates the effects of the polymorphism has a protective role [968] . Kinins indeed bind to Gq-coupled bradykinin B 1 and B 2 receptors to reduce the risk of diabetic complications. In adult rat cardiomyocytes, bradykinin activates P21 activated kinase PAK1 downstream from monomeric GTPases. Kinase PAK1 stimulates protein phosphatase-2 that dephosphorylates both cardiac troponin-I and phospholamban [971] . Protein phosphatase-2 is able to complex with both PAK1 and phospholamban. Bradykinin is a potent vasodilator of arteries that irrigate muscle, kidney, and heart, among other viscera. Tissue kallikrein does not contribute to uterine artery remodeling during and after pregnancy [972] . Bradykinin synthesized by tissue kallikrein from kininogen stimulates endothelial β2-adrenergic receptors, subsequently releasing nitric oxide 101 and prostacyclin to control the vascular tone. Both receptors are coupled with Gq proteins. Their stimulation activates phosphatidylinositol-specific phospholipases that augment intracellular Ca ++ concentration and leads to activation of endothelial nitric oxide synthase (NOS3). Kinins that act via B 2 also support the expression of inducible NOS (NOS2). Nitric oxide reversibly suppresses mitochondrial oxidation, at least partly by inhibition of cytochrome-C oxidase of the electron transport chain. Kinins also facilitate the synthesis of prostanoids, such as prostaglandins PGE2 and PGI2, which heighten intracellular cAMP concentration [968] . Messenger cAMP also decreases mitochondrial respiration, as it activates the NADH-ubiquinone oxidoreductase activity of complex I and inhibits cytochrome-C oxidase. Therefore, nitric oxide and kinins reduce oxidative stress. In particular, bradykinin reduces mitochondrial superoxide generation in human vascular endothelial cells [968] . Bradykinin primes afferent reflex in the heart owing to cardiac sympathetic afferent fibers. Bradykinin causes a sympathoexcitatory reflex 102 characterized by 100. This polymorphism does not significantly affect blood pressure and angiotensin-2 and aldosterone concentration. Nevertheless, it is associated with different risks of diabetic complications. 101. Endothelial nitric oxide synthase expression and, consequently, nitric oxide production, increase during pregnancy. 102. The sympathoexcitation reflex happens when all other cardiovascular reflexes (baroreceptor reflex, cardiopulmonary reflex with vagal afferents) are not efficient enough. increased arterial blood pressure and renal sympathetic nerve activity via epicardial B 2 receptors and sympathetic cardiac afferent fibers [973] . Intrapericardial bradykinin generates a blood pressure rise, whereas circulating bradykinin acts as a vasodilator, thereby lowering blood pressure. In the heart, bradykinin is released during cardiac ischemia and myocardial infarction. Both bradykinin receptors B 1 and B 2 contribute to the cardioprotective effect of ACE inhibition mediated by bradykinin [974] . The B 2 receptor is the main kinin receptor involved in the cardioprotection yielded by ischemic preconditioning [975] . It forms a complex with angiotensin-converting enzyme that sequesters the latter. This process determines a crosstalk between the renin-angiotensin pathway and the kinin-kallikrein axis. Calcitonin (Ct) 103 is a hormone that results from the proteolytic cleavage of a prepropeptide that is encoded by the CALCA gene (or CALC1) primarily in parafollicular cells 104 of the thyroid. Procalcitonin (PCt) belongs to a family of related hormone precursors that include islet amyloid precursor, calcitonin gene-related peptide, and adrenomedullin precursor. It decreases bone resorption. Amylin (Amy) 105 is a hormone secreted by pancreatic β cells. Amylin reduces nutrient intake and contributes to the control of glycemia, as it optimizes glucose metabolism. Calcitonin gene related peptide-2 (CGRP2) 106 is a member of the calcitonin family of peptides. It is produced in both peripheral and central neurons. It is a potent vasodilator. Adrenomedullin-1 (AM1) is ubiquitously synthesized from a precursor (proadrenomedullin) encoded by the AM gene. It abounds in vascular endothelial and smooth muscle cells. It is also a potent vasodilator. Furthermore, it regulates body fluids, as it precludes drinking and sodium intake and causes natri-and kaliuresis [976] . 107 Different excisions form various peptides, such as an inactive adrenomedullin, ePAMP, adrenotensin, and AM 95−−146 . In humans, mature adrenomedullin is rapidly catabolized. Circulating adrenomedullin comprises both mature, amidated and inactive, glycated form. The related peptide adrenomedullin-2 (AM2), or intermedin, participates in the regulation of blood circulation. It raises the concentrations 103. A.k.a. calcitonin-related polypeptide-α, CalcA, and Calc1, as well as calcitonin generelated peptide CGRP1 and α-CGRP (Vol. CL-RAMP3 Gs AM1 ≥ CGRP, AM2 > Amy of atrial and brain natriuretic peptides and renin in plasma. Adrenomedullin-5 that also derives from a prohormone decreases arterial pressure. Calcitonin, amylin, calcitonin gene-related peptide, and adrenomedullin receptors are generated by the genes CALCR and CALCRL that encodes the calcitonin receptor (CalcR, CTR, and CTR1) and calcitonin receptor-like receptor (CalcRL and CLR). Calcitonin receptors that activate G-protein subunits Gs and Gq are involved in the maintenance of calcium homeostasis. Their function changes according to their interaction with receptor activity-modifying proteins (RAMP1-RAMP3) that form multimeric amylin receptors Amy 1 (CalcR-RAMP1), Amy 2 (CalcR-RAMP2), and Amy 3 (CalcR-RAMP3; Table 7 .47). Splice variants of the CalcR receptor produce variants of Amy receptors. Calcitonin receptor-like receptors also connect to receptor activity-modifying proteins. Receptor of CGRP2 (CGRPR) is a heteromer composed of the G-proteincoupled receptor CalcRL and receptor activity-modifying protein RAMP1. The Cal-cRL subunit also produces the adrenomedullin receptor AM 1 and AM 2 with RAMP2 and RAMP3, respectively. Adrenomedullin-1 targets the CalcRL-RAMP2 or CalcRL-RAMP3 complexes, whereas adrenomedullin-2 interacts non-selectively with all 3 CalcRL-RAMP heteromers [978] . Adrenomedullin-5 tethers to AM 5 receptors (other than CalcRL-RAMP or CalcR-RAMP) to exert its cardiovascular actions in mammals [979]. Calcium-sensing receptors (CaR, CaS, or CaSR) are G-protein-coupled receptors (Table 7 .48) that sense the concentration of extracellular Ca ++ ions. Calciumsensing receptor responds to extracellular concentrations of calcium and magnesium in the millimolar range and of gadolinium and some polycations in the micromolar range [5] . The extracellular Ca ++ -sensing receptor responds also to many other ligands. In particular, its activity is allosterically regulated by amino acids and H + ions. This GPCR resides especially on hematopoietic stem cells. It retains hematopoietic stem cells close to the endosteal surface of the bone marrow. It interacts with extracellular matrix components, particularly collagen-1. Calcium-sensing receptor also lodges in parathyroids, where it participates in parathyroid hormone response to extracellular Ca ++ concentration ([Ca ++ ] e ). In addition, CaRs are produced by cells of the kidney, liver, and thyroid gland. In the liver, calcium-sensing receptor is specifically expressed in hepatocytes (not in stellate, endothelial, and Kupffer cells). It mobilizes calcium ions from IP 3sensitive stores to stimulate bile flow [980] . This major sensor and regulator of [Ca ++ ] e also localizes to the stomach and intestinal tract, especially on gastrin-secreting G cells in the stomach. Luminal nutrients, particularly Ca ++ and amino acids are potent stimulators of gastrin and acid secretion [981] . In cardiomyocytes, CaSR predominantly localizes to caveolae. It ensures a cardioprotective role in ischemic preconditioning [982] . Cannabinoid receptors are targeted by endo-and exogenous cannabinoids. Endocannabinoids comprise anandamide, or N arachidonoyl ethanolamine (AEA), as well as N homo γ-linolenoyl ethanolamine, N docosatetra (7,10,13,16)-enoyl ethanolamine, and 2-arachidonyl glycerol (2AG). 108 Anandamide and 2-arachidonoyl glycerol are 2 major endocannabinoids. Endocannabinoids 2-arachidonyl glycerol and anandamide are released from the cell membrane by activated phospholipases. Cyclooxygenase-2 transforms 2-arachidonyl glycerol and anandamide into prostaglandin glycerol esters ( PGH2 G) and ethanolamides ( PGH2 EA). 109 In cells, glycerol esters and ethanolamides of prostaglandins PGE2, PGD2, and PGF2α are endocannabinoid-derived COx2 products ( PGH2 G and PGH2 EA). The sequential action of COx2 and thromboxane-A synthase (TxaS) on AEA and 2AG yields thromboxane-A2 ethanolamide and glycerol ester, respectively [983] . Similarly, prostaglandin synthases PGdS, PGeS, and PGiS (i.e., prostaglandin-D, -E, -H2, and prosta-108. 2-Arachidonyl glycerol is also termed 2-arachidonoyl-glycerol, (5,8,11,14)eicosatetraenoic acid, and 2-hydroxy 1-(hydroxymethyl)ethyl ester. 109. Activated phospholipase releases arachidonic acid from phospholipids. Afterward, cyclooxygenase oxygenates liberated fatty acid to generate the hydroxy-endoperoxide prostaglandin-H2. The latter can be converted into one among various derivatives: prostaglandins, thromboxane, and prostacyclin. Thromboxane-B2 (TxB2) is an inactive TxA2 metabolite. Cyclooxygenase-1 generates thromboxane-A2 in platelets. cyclin synthases) contribute to the formation of prostaglandin and prostacyclin glycerol esters and ethanolamides. The endocannabinoid system is involved in signal transduction not only in the central nervous system, but also in other organs. Endocannabinoids are lipid mediators that are released and rapidly degraded by numerous enzymes. The most abundant endocannabinoid in the central nervous system, 2-arachidonoyl glycerol, is synthesized from diacylglycerol by diacylglycerol DAGLα (mainly) and DAGLβ lipases. These enzymes may also produce arachidonic acid. 2-Arachidonoyl glycerol functions as a retrograde signaling molecule that suppresses synaptic transmission in the central nervous system and regulates axonal growth and guidance. 110 Two known cannabinoid isoreceptors are: (1) type-1 (CB 1 ) expressed mainly in the brain, but also lung, liver, and kidneys, as well as vascular smooth muscle and endothelial cells, monocytes, and macrophages; and (2) type-2 (CB 2 ) cannabinoid receptor chiefly produced in hematopoietic cells (B and T lymphocytes as well as macrophages), and also keratinocytes and vascular smooth muscle and endothelial cells. Novel cannabinoid receptors exist in endothelial cells as well as in the central nervous system. Cannabinoid-like receptors encompass GPR18 for N arachidonoylglycine, GPR55 with a wide spectrum of cannabinoid agonists and antagonists, GPR119 for fatty acid ethanolamides [5] . Anandamide also targets TRPV1 channel. Cannabinoid receptors participate in the regulation of adult neurogenesis [984]. Receptors CB 1 and CB 2 participate in the genesis of atherosclerosis. Monocytes undergo a significant change in CB 1 and CB 2 expression profile during differentiation into macrophages [985] . The CB 1 :CB 2 ratio on monocytes and macrophages, indeed, regulates their inflammatory activity and ability to produce reactive oxygen species [985] . These receptors have opposing influences on ROS production. In macrophages, CB 1 , but not CB 2 , causes phosphorylation of P38MAPK that causes ROS production and synthesis of pro-inflammatory cytokines such as tumor-necrosis factor-α and CCL2 chemokine. 111 On the other hand, the CB 2 receptor activates small GTPase Rap1 that counteracts CB 1 -stimulated ROS production. In endothelial cells, the CB 2 receptor also lowers TNFα-induced proliferation and migration of smooth muscle cells and expression of adhesion molecules and chemokines. The type of cannabinoid receptor ligand influences Gα-subunit response. A given ligand of cannabinoid receptor CB 1 can act as an agonist for Gi1 and Gi2 and as an antagonist for Gi3. Another type of ligand can behave as an inverse agonist of Gi1 and Gi2 and as an agonist for Gi3. Cannabinoid receptors target many effectors, such as adenylate cyclase, inwardly rectifying potassium channels (K IR ) and other K + channels, calcium channels, protein kinase-A and -C, kinases Raf1, ERK, JNK, and P38MAPK, and transcription factors Fos and Jun. Endocannabinoids released from a neuron that bears endocannabinoid action bind to CB 1 receptor in presynaptic neuron and reduces GABA liberation, hence GABA-mediated neurotransmission. Cannabinoid receptor CB 1 targeted by glucocorticoids in the basolateral nucleus of the amygdala intervenes in the consolidation of emotional memory, although glucocorticoids usually regulate gene transcription by either binding homo-or heterodimers of intracellular glucocorticoid receptors to nuclear DNA or by interacting with transcription factors [986] . Endogenous cannabinoid binding to CB 1 stimulates appetite. Activated presynaptic CB 1 receptors also repress sympathetic innervation of blood vessels. Anandamide and 2-arachidonyl glycerol produced by macrophages and platelets, respectively, can cause hypotension. In the liver, activated CB 1 increases fatty acid synthesis via lipogenic transcription factor SREBP1c and acetyl coenzyme-A carboxylase-1 and fatty acid synthase [987] . Cannabinoid CB 2 receptor abounds in mature B lymphocytes. It also exists in myeloid and natural killer cells, and various other cell types. Ligand for CB 2 , 2arachidonoyl glycerol, is generated from arachidonic acid-containing phospholipids. Expression of CB 2 in peripheral B lymphocytes is correlated to the production level of immunoglobulin λ-chain [988] . Production and function of CB 2 are upregulated in immature B lymphocytes. Migration in bone marrow sinusoids of immature B cells depends on Gi-proteincoupled cannabinoid receptor CB 2 [988]. Immunoglobulin-M+ immature B cells can be retained in bone marrow sinusoids owing to integrin-α 4 β 1 and its endothelial ligand VCAM1. G-protein-coupled receptor GPR119 can also serve as a cannabinoid receptor. It is synthesized predominantly in pancreas and gastrointestinal tract. It regulates the secretion of incretin and insulin. It is activated by endogenous fatty acid ethanolamines, particularly N oleoylethanolamine and N palmitoylethanolamine [5] . It is mainly coupled to Gs subunit. Chemokine receptors constitute a large class of GPCRs that are activated by at least one chemokine. 112 Chemokines can be subdivided according to their structure into 4 subclasses: 28 known CC, 113 16 identified CXC, 114 2 detected C, and 1 CX 3 C chemokines. Chemokines can also be classified according to their function into homeostatic and inflammatory groups. Most chemokine receptors can bind with a high affinity to multiple chemokines (Table 7 .49, 7.50, and 7.51). Most chemokines tether to several receptor subtypes. However, chemokines bind almost always to the same structural receptor subclass. Chemokine receptor CCR7 expressed on mature, naive T lymphocytes intervenes in immune responses that require the coordinated interaction of various cell types 112. I.e., cytokine with chemotactic activity for leukocytes. 113. A.k.a. β-chemokines. 114. A.k.a. α-chemokines. within lymphoid tissues. Agonists CCL19 and CCL21 target CCR7 to initiate chemotaxis. Ligand CCL19 leads to CCR7 phosphorylation by both GRK3 and GRK6 and β-arrestin-2 recruitment, whereas CCL21 activates only GRK6. Only CCL19 leads to receptor desensitization, internalization, and degradation, whereas both agonists launch extracellular signal-regulated protein kinase via GRK6 [989]. The Gi-coupled chemokine receptor CXCR4 binds to CXCL12 produced by bone marrow stromal cells. It enables the retention of neutrophils and hematopoietic stem and progenitor cells in the bone marrow. 115 Conversely, disruption of CXCL12mediated chemoattraction of CXCR4+ cells mobilize neutrophils and hematopoietic stem and progenitor cells into the blood circulation. A pepducin 116 mobilizes bone 115. The CXCR4 receptor resides on hematopoietic stem cells, myeloid progenitors, and immature neutrophils. 116. A pepducin is composed of a peptide derived from the amino acid sequence of one of the intracellular loops of a target GPCR coupled to a lipid. Synthetic pepducins are thus cell- . It can be thus used to more easily collect hematopoietic stem and progenitor cells before autologous bone marrow transplantation. Anaphylatoxins, or anaphylotoxins, are fragments (C3a, C4a, and C5a) produced once the complement cascade is activated. Anaphylatoxins are able to trigger degranulation of endothelial and mastocytes and phagocytes to produce a local inflammation. Anaphylatoxins indirectly cause smooth muscle cell contraction, increase blood capillary permeability, and provoke chemotaxis of leukocytes. Anaphylatoxin and formyl peptide receptors are activated by anaphylatoxins C3a and C5a (Table 7 .52). penetrating lipopeptides. Any subtype of pepducins inhibits signal transmission from GPCRs to G proteins, Table 7 .52. Receptors of the complement system and anaphylatoxin and formyl peptide receptors, targeted G protein subunits, and order of potency (Source: [5] ; fMLP: formyl-Met-Leu-Phe). Gi/o, Gi/z C3a > C5a C5aR Gi/o, Gi/z, Gq/16 C5a > C3a FPR1 Gi/o, Gz fMLP > cathepsin-G > annexin-1 Formyl peptide receptors (FPR) 117 are expressed at high levels on phagocytes. They are involved in chemotaxis, production of reactive oxygen species, and release of peptidases. Activated formyl peptide receptors trigger cytoskeleton rearrangement that, in turn, facilitates cell migration. They lead to activation of: (1) phospholipase-C and the IP 3 -Ca ++ and DAG-PKC cascades; (2) small GTPase Ras and the MAPK module; and (3) CD38 ectoenzyme that converts NAD + into cyclic ADP ribose, which interacts with ryanodine receptors for sustained Ca ++ influx. In addition, formyl peptide receptor-like proteins of vomeronasal sensory neurons have an olfactory function associated with pathogens. Cholecystokinin receptors (CCK 1 -CCK 2 or CCKa-CCKb) belong to the class A of G-protein-coupled receptors. They are activated by cholecystokinins Cck4, Cck8, and Cck33, as well as gastrin (Table 7 .53). The CCK 2 receptor is the dominant receptor in the central nervous system [991]. It is highly synthesized in the gastric mucosa, where it mediates gastrin-stimulated gastric acid secretion. It also contributes to gastric epithelial renewal. It resides in exo-and endocrine pancreas, enteric smooth muscle, liver, kidney, and adrenal gland, as well as circulating monocytes. The CCK 2 receptor recognizes each of the mature forms of cholecystokinins and gastrin with similar affinities and responds to them with similar potencies. It binds to progastrin and glycine-extended gastrin with lower affinity. It can couple to Gq G-protein subunit to signal predominantly via phospholipase-C and calcium import. It can also couple to Gs subunit. It 117. Formyl peptide receptors have been originally identified by their ability to bind N formyl peptides, such as exogenous bacterial product, the formylated tripeptide N formyl-methionylleucyl-phenylalanine (fMLP). The fMLP receptor is now called FPR1. Corticotropin-releasing factor receptors CRF 1 (or CRFR1) and CRF 2 (or CRFR2) are activated by peptides of the CRF family, i.e., CRF 118 and urocortins (Ucn1-Ucn3; Table 7 .54). 119 Urocortin-1 connects to both CRF 1 and CRF 2 , whereas Ucn3 is a high-affinity ligand for CRF 2 receptor. Activation of CRF 2 on afferent terminals in the medial nucleus tractus solitarius by Ucn1 and Ucn3 releases glutamate that, in turn, causes a decrease in mean arterial pressure and cardiac frequency via activation of ionotropic glutamate receptors [993] (Sect. 2.5.4). Corticotropin-releasing factor is a major neuroregulator of the hypothalamuspituitary-adrenal axis that has many central and peripheral actions. The CRF 1 receptor is expressed in anterior pituitary corticotropes. In response to hypothalamic CRF hormone, CRF 1 triggers the release of adrenocorticotropic hormone. This release causes the secretion of glucocorticoids from the adrenal cortex that, in particular, stimulates liver gluconeogenesis to increase glycemia. Activity of CRF 1 primed by CRF complements the action of CRF 2 receptor activated by urocortin-3 that is responsible for auto-and paracrine glucose-mediated stimulation of insulin secretion. In pancreatic β cells that produce urocortin-3, activation of CRF 1 promotes insulin secretion [994] . Therefore, the insulinotropic action of pancreatic CRF 1 antagonizes the effect of activated CRF 1 on anterior pituitary corticotropes, as the release of glucocorticoids counteracts the action of insulin. 118. A.k.a. corticotropin-releasing hormone (CRH). 119. Urocortin-2 and -3 are also called stresscopin-related peptide and stresscopin, respectively. Urocortin-1 and -3 are members of the corticotrophin-releasing factor (CRF) peptide family. The CRF 1 receptor stimulates insulin secretion only when glucose concentration is intermediate to high. The higher the glycemia, the greater the CRF 1 -dependent phosphorylation of extracellular signal-regulated protein kinases ERK1 and ERK2 and subsequent cAMP response element-binding phosphorylation. Phosphorylation of ERK1 and ERK2 caused by CRF is similar to glucose-dependent actions of incretins, glucagon-like peptide GLP1 and glucose-dependent insulinotropic peptide (GIP), via incretin receptors GLP1R and GIPR that pertain to the class-B G-proteincoupled receptors. Dopamine is the predominant catecholamine neurotransmitter in the central nervous system as well as a neurohormone released by the hypothalamus to inhibit the release of prolactin from the adenohypophysis (anterior lobe of the pituitary gland). 120 Dopamine receptors include 5 proteins (D 1 -D 5 ) distributed in 2 groups: (1) the D 1 -like (D 1 and D 5 ) and (2) D 2 -like (D 2 -D 4 ) groups (Table 7 .55). Two D 1 -like receptor subtypes couple to Gα s subunit, thereby activating adenylate cyclase. The other receptor subtypes of the D 2 -like group inhibit adenylate cyclase via Gα i/o and activate K + channels. The D 1 and D 5 receptor genes are intronless. Pseudogenes of D 5 exist. Alternative splicing yields different species-and tissue-dependent types of D 2 and D 3 receptors. In humans, the D 4 receptor gene exhibits extensive polymorphic variation [995] . In the central nervous system, dopamine receptors are involved in the control of locomotion, cognition, emotion, affect, food intake, and neuroendocrine secretion. In peripheral organs, dopamine receptors are prominent in the vasculature, kidney, and pituitary gland, where they influence mainly sodium homeostasis, vascular tone, renal function, gastrointestinal motility, and hormone secretion [995] (Table 7 .56). 120. The adenohypophysis regulates stress response, growth, and reproduction. The Fos transcription factor, a product of the immediate-early FOS gene required for long-lasting modifications of gene expression in response to acute stimuli, is a final targets in the signaling initiated by dopamine receptors. The D 1 and D 2 receptors operate synergistically on FOS expression [995]. Receptor D 1 stimulates phospholipase-C (PLC), thereby leading to the production of inositol trisphosphate that causes Ca ++ influx by mobilization of calcium from intracellular stores on the one hand, and activating PKC enzyme. Receptor D 1 can stimulate the activity of Ca V 1 channel via the cAMP-PKA pathway [995] . On the other hand, D 1 reduces Ca ++ flux through Ca V 2.1 and Ca V 2.2 channels due to channel dephosphorylation by a phosphatase stimulated by PKA enzyme. Receptor D 1 precludes the activity of Na + -H + exchanger that regulates intracellular pH and cell volume using both cAMP-dependent and -independent mechanisms [995] . Moreover, D 1 inhibits Na + -K + ATPase that maintains the electrochemical gradient for appropriate excitability of neurons and myocytes and enables the transport of fluid and solutes across epithelial membranes [995] . However, in the kidney, dopamine receptor activates Na + -K + ATPase via phosphorylation by PKA and PKC kinases. In the kidney, dopamine is produced in renal nerves and epithelial cells of certain nephron segments. Dopamine synthesized in the renal tubular epithelium acts as a para-and autocrine factor that regulates sodium reabsorption in the nephron. In addition, dopaminergic nerve endings lodge near the juxtaglomerular apparatus. Receptor D 1 stimulate the secretion of renin [995] . Dopamine impedes Na + -K + AT-Pase via both D1-like and D2-like receptors (D 1 -D 2 synergism). Both D 1 and D 2 receptors in the hippocampus mediate the effect of dopamine on learning and memory [995] . Both D 1 and D 2 receptors are involved in mesolimbocortical effect (reward and reinforcement) of dopamine. Striatal efferent neurons undergo the influence of dopamine. Two major types of neurons are identified by their primary sites of axonal projections and neuropeptide synthesis [995] . A first striatonigral population projects to the entopeduncular nucleus and substantia nigra pars reticulata and synthesizes substance-P and dynorphin, an opioid peptide stored in large (size 80-120 nm), dense-core vesicles. Striatonigral gabaergic neurons preferentially express D 1 receptor that fosters the production of substance-P and dynorphin. The second striatopallidal pool projects to the external segment of the globus pallidus and contains enkephalin. Striatopallidal gabaergic neurons prominently synthesize D 2 receptor that prevents the production of preproenkephalin. A similar organization of dopamine receptors is observed in the nucleus accumbens: D 1 receptor is mostly expressed in substance-P+ neurons, D 2 receptor in enkephalin+, neurotensin+ neurons, and D 3 receptor in substance-P+, neurotensin+ neurons [995] . However, in most of the neuronal populations, D 1 and D 2 are coexpressed rather than segregated. In the cardiovascular system, more precisely in blood vessels, postjunctional D 1 receptor provokes vasodilation of the renal artery and prejunctional D 2 receptor on postganglionic sympathetic nerve terminals prevents noradrenaline release, thereby indirectly causing vasodilation in the femoral artery [995]. Two alternatively spliced D 2 isoforms exist (D 2 L and D 2S ) [995] . Both isoforms may prime a phosphatidylinositol-mediated mobilization of intracellular calcium. However, in many other cell types, D 2 receptor do not launch this pathway. It can also inhibit inward calcium currents. In addition, D 2 receptor increase outward potassium currents, thareby provoking a cell hyperpolarization. Receptor D 2 potentiates the release of arachidonic acid via Gi subunit [995] . Unlike D 1 , D 2 receptor activates Na + -H + exchanger in many cells. The D 2 dimer binds to a single heterotrimeric G protein [996] . The maximal activation of the minimal signaling unit, i.e., a receptor dimer coupled to a single heterotrimeric G protein, is achieved by agonist binding to a single subunit of the dimeric D 2 receptor. The asymmetrically activated dimer can be modulated by the activity state of the second monomer. Ligand-independent constitutive activation of and inverse agonist binding to the second monomer enhances signaling. In the ventral striatum, activated D 2 autoreceptor reduces dopamine release, hence locomotor activity [995] . On the other hand, activated postsynaptic D 2 receptor slightly increases locomotion. Receptors of the D 2 -like group may promote some aspects of cell differentiation [995] . Prejunctional D 2 receptor on postganglionic sympathetic nerve terminals attenuates the cardiac contractility [995] . Dopamine represses aldosterone secretion by adrenal glomerulosa cells via D 2 receptor [995] . During sodium depletion, dopamine release decays and circulating aldosterone level and plasma aldosterone responsiveness to angiotensin-2 augments. Dopaminergic neurons of sympathetic ganglia inhibit release of adrenaline and noradrenaline by neuroendocrine chromaffin cells of the medulla of adrenal glands via D 2 receptor [995] . Adrenal chromaffin cells possess also D 1 receptor. The latter causes a rapid catecholamine secretion in response to stress. Receptor D 3 localizes mainly postsynaptically to the nucleus accumbens, where it impedes locomotion, hence antagonizing effect of D 2 receptors [995]. Receptor D 3 have an opposite action w.r.t. that of D 2 on neurotensin gene expression in the nucleus accumbens. 121 Receptor D 3 stimulates thymidine incorporation in some cell types [995] . It may activate the mitogen-activated protein kinase module. Different forms of the D 4 receptor exist with different number of repeats in the third intracellular loop. The four-repeat form (D 4 4 ) is the predominant in the human population (60%) [995]. 122 Receptor D 5 is highly expressed in the hippocampus, where it may mediate the effects of dopamine on learning and memory [995] . It is coexpressed with D 2 in cholinergic interneurons of the striatum. 121 . In the ventral shell of the nucleus accumbens, D 3 receptor activates neurotensin synthesis. In the septal pole of the nucleus accumbens, D 2 receptor hinders neurotensin production. 122. The D 4 7 variant is observed in 14% of the population and the D 4 2 in 10%. Endothelin is secreted by endothelial cells and targets neighboring smooth muscle cells to regulate the vasomotor tone (Table 7 .57; Vol. 5 -Chaps. 8. Smooth Muscle Cells and 9. Endothelium). Among 4 identified isoforms (ET1-ET4), endothelin ET1 is the most commonly found subtype in the cardiovascular system, including blood. Endothelins are synthesized by vascular endothelial cells of both vessel (intima) and heart (endocardium) walls (especially endothelial cells of right atrium and left ventricle), vascular smooth muscle cells, and cardiomyocytes, as well as by extravascular cells of the lung, pancreas, spleen, and nervous system [5] . Vasoconstrictor endothelin-1, the major type in the vasculature, intervenes as an auto-and paracrine regulator. Endothelin-1 indeed either counteracts (paracrine action on vascular smooth muscle cells) or cooperates with vasodilator nitric oxide (via an autocrine loop on endothelial cells). Physical factors such as wall shear stress and chemical agents, such as thrombin, adrenaline, angiotensin-2, growth factors, cytokines, and free radicals trigger ET1 secretion. Several substances, such as nitric oxide, cGMP, atrial natriuretic peptide, and prostacyclin reduce ET1 release from the vascular endothelium. Two endothelin-1 receptors (ET A and ET B ) 123 bind ET1 with equal affinity to regulate the gene expression devoted to cell contraction, proliferation, and survival. Receptor subtypes ET A and ET B include ET A1 and ET A2 and and ET B1 and ET B2 . The vasoconstrictor activity of endothelins is mediated by Na + -H + and Na + -Ca ++ exchangers. An initial activation of Na + -H + exchangers causes an increase in Na + concentration inside the smooth muscle cell [997] . Subsequently, Na + -Ca ++ exchangers export Na + and import Ca ++ into the smooth muscle cell that provokes contraction. The Na + -H + exchanger also reduces the intracellular H + concentration. Messengers ET1 to ET3 potentiate the mitogenic activity of platelet-derived growth factor via ET A receptors. Endothelin-1 is a potent mitogen for both vascular endothelial and smooth muscle cells. Ligand ET1 activates extracellular signalregulated kinase ERK1 and ERK2 by GPCR-induced activation of the receptor Tyr kinase EGFR (Sect. 8.2.5.2). 124 123. A third receptor subtype specific for ET3 isoform -ET C -exist in Xenopus laevis, but not in mammals. 124. G-protein-coupled receptors can indeed activate Src kinases with subsequent RTK phosphorylation. Once activated, Src and/or RTK phosphorylate SHC adaptor. Both phosphorylated SHC and activated RTK then bind growth factor receptor-bound protein GRB2 and Type-A endothelin-1 receptor phosphorylates the docking growth factor receptorbound protein-2 (GRB2)-associated binder-1 (GAB1) and activates ERK1 [998] . 125 Phosphorylation of GAB1 induced by ET1 is stimulated by phosphoinositide 3kinase and Src kinases and inhibited by PP1 phosphatase. Protein Tyr phosphatase PTPn11 potentiates ET1-induced ERK1 activation. Receptor ET B localizes to the nervous system, heart, vasculature, lung, endocrine system, liver, gastrointestinal tract, pancreas, and placenta [999] . In the cardiovascular apparatus, ET B is mainly produced in endothelial cells; it is synthesized at low levels in vascular smooth muscle cells and cardiomyocytes. It can lodge in caveolae. igands ET1, ET2, and ET3 activate ET B with equal affinity and potency. Among 3 splice variants, variant-1 is the canonical isoform. Variant-1 and -3 have functional characteristics; variant-2 that has similar binding properties, but lacks the functional coupling to phosphoinositide [999] . Both ET B and ET A can homo-and heterodimerize. In addition, ET B can heterodimerize with dopamine D 3 receptor and angiotensin AT 1 receptor in rat renal proximal tubule cells [999] . Most ET B effects result from coupling to Gq and Gi, but ET B can also couple to other G-protein subunits, such as Go, Gs and G12/13. Receptor ET B is thus involved in the inhibition or activation of adenylate cyclase, activation of phospholipase-A2, -C, and -D, guanylate cyclase, protein kinases such as mitogen-activated protein kinases [999] . In vascular smooth muscle cells, liganded ET B causes a biphasic activation of ERK1 and ERK2 kinases. The delayed phase of ERK activation follows ET B proteolysis by metallopeptidase and EGFR transactivation. Two ET B subtypes may reside on endothelial and vascular smooth muscle cells to cause vasodilation and vasoconstriction, respectively. In any case, activated ET B counteracts the potent vasoconstrictor effect of ET A stimulation, as it provokes vasodilation, natriuresis, and clearance of ET1 from the circulation by endothelial cells of the lung, kidney, and liver. However, ET B is produced at low concentrations in vascular smooth muscle cells, in which it causes contraction [999] . In the lung, ET B on airway smooth muscle cells launches bronchoconstriction and cell proliferation [999] . Receptor ET B can be phosphorylated (deactivated) by G-protein coupled receptor kinase GRK2 [999] . The agonist-ET B complex, like liganded ET A , dissociates very slowly. Following ligand binding, ET B receptor is internalized using a dynamin-clathrin-dependent pathway for lysosomal degradation. Endocytosis of the its associated Ras guanine nucleotide-exchange factor Son-of-sevenless to trigger the Ras-MAPK signaling. 125. The docking protein GAB1 can be phosphorylated by receptor Tyr kinases and cytokine receptors to regulate cell survival and proliferation aa well as from activated G-protein-coupled receptors. In the coronary bed of rat heart, ET A is detected in arterial smooth muscle and capillary endothelial cells, whereas ET B resides in arterial, venous, and capillary endothelial cells as well as arterial and venous smooth muscle cells [1000] (Table 7 .58). Receptors ET A and ET B control arterial vasoconstriction. Postcapillary vascular resistance is exclusively regulated by ET B . The presence of ET A in capillary endothelium attenuates increase in vasomotor tone, especially in cardiac microvascular permeability during ischemia-reperfusion events. Cultures of human coronary and internal mammary arteries, as well as rat omental, mesenteric, and cerebral arteries during 24 to 48 h modify endothelin receptor expression on endothelial and smooth muscle cells. Both endothelin receptor types are upregulated in vascular smooth muscle cells in vessel culture. Conversely, ET B s are downregulated in endothelial cells in vessel culture [1001] . Ligand administration during 5 d in dogs induces a heterogeneous distribution of endothelin receptors in coronary arteries [1002] . Right coronary artery is enriched in ET B 3 times more than left coronary artery (in vascular smooth muscle cells). Endothelin-1 increases the myocardial contractility (positive inotropic effect) with a greater potency than endothelin-3. Receptor ET A is involved in ET-induced increase in myocardial contractility. Activated ET B receptor has an opposite effect [997] . However, ET B receptor can lead to positive inotropic effect via coronary vasodilation that increases myocardial contraction and oxygen consumption (Gregg's phenomenon). 126 126. Coronary perfusion pressure affects coronary arterial resistance (autoregulation) as well as myocardial oxygen consumption (Gregg's phenomenon). Gregg's phenomenon results from increased contractility. Signal transduction relies on 2 intertwined processes: (1) signal transmission by stimulating appropriate mediators and (2) signaling termination to avoid prolonged reaction and prepare cells for forthcoming stimulation. G-Protein-coupled receptor kinases (GRK1-GRK7) enable a rapid GPCR desensitization after activation. In vascular smooth muscle cells, the predominant ET1 receptor ET A is desensitized by GRK2 [1003]. Endothelin receptors ET A and ET B have opposite roles (Table 7 .59). The ET B receptor mediates vasodilation 127 as well as endothelial cell survival and proliferation. 128 On the other hand, the ET A receptor favors vasoconstriction. Responses to ET B signaling depends on cell types. Ablation of ET B exclusively from endothelial cells decreases NO release and increases plasma endothelin-1 [1006]. Endothelial ET B receptor mediates vasodilation without affecting blood pressure in response to a high-salt diet, whereas non-endothelial ET B controls blood pressure. The amount of circulating endothelin-1 and endothelial nitric oxide synthase increases and decreases in persistent pulmonary hypertension, respectively. Endothelin-1 increases ROS production in pulmonary arterial smooth muscle cells in culture. Reactive oxygen species downregulate NOS3 in smooth muscle cells [1007] . Endothelin-1 increases H 2 O 2 level in fetal pulmonary arterial smooth muscle cells via ET A in vitro. On the other hand, ET1 decreases H 2 O 2 level in fetal pulmonary arterial endothelial cells in monoculture via ET B receptor. Furthermore, H 2 O 2 at low concentration (12 μmol) increases NOS3 level without affecting NOS3 promoter activity, whereas at higher concentration (100 μmol) it reduces both NOS3 promoter activity and NOS3 level in fetal pulmonary arterial endothelial cells in monoculture. 127. The ET B receptor is functionally coupled to NOS to produce NO via a Tyr kinase-and calcium-calmodulin-dependent pathway [1004] . 128. Endothelin-1 promotes the proliferation and migration of human umbilical vein endothelial cells in a dose-dependent manner, by activating ET B more effectively than ET A [1005] . In addition, ET1 stimulates matrix metallopeptidase-2 production. It also enhances VEGF angiogenic effects on endothelial cells in vitro. In addition, ET1 decreases NOS3 promoter activity in these cells cultured with fetal pulmonary arterial smooth muscle cells. Endothelin is able to exert: (1) a persistent constrictor effect by increasing the intracellular calcium concentration resulting from an influx through activated Na + -H + and Na + -Ca ++ exchangers and from cellular stores due to the stimulated PLC-IP 3 pathway in vascular smooth muscle cell and (2) a transient dilator effect via the activation of endothelial nitric oxide synthase mediated by protein kinase-B and receptor ET B1 [997] . Receptor subtypes ET B1 (endothelial) and ET B2 (muscular) mediate opposite effects on vascular tone. Receptors ET A and ET B2 indeed prime vasoconstriction, whereas receptor ET B1 triggers transient vasodilation. Endothelin-1 intervenes during birth when pulmonary vascular resistance falls with the initiation of ventilation and the ductus arteriosus constricts, as well as postnatally. At birth, in both term and preterm, oxygen triggers long-lasting vasoconstriction and closure of ductus arteriosus by activating a specific, cytochrome-P450mediated reaction that leads to ET1 synthesis; ET1 then operates via predominant constrictor ET A as well as ET B2 [1009] . The number of ET B1 dilator receptors is reduced in pulmonary hypertension. Receptor ET A is able to increase myocardial contractility. This inotropic effect results from an increase in intracellular Ca ++ concentration induced by Na + -H + and Na + -Ca ++ exchangers stimulated by protein kinase-C, which is activated by the PLC-DAG axis initiated by endothelin. Receptors ET B1 and ET B2 may mediate negative and positive inotropic effects, respectively [1008]. Endothelin-1 is a potent cardiomyocyte survival factor. It causes nuclear translocation of PP3-regulated nuclear factors of activated T cells NFAT1 and production of anti-apoptotic BCL2 protein. Factor P300 potentiates NFAT1 binding to the Bcl2 gene promoter in cardiomyocytes [1010] . Transcription factor NFAT also regulates transcription of several genes, such as those that encode adenylosuccinate synthase AdsS1, heart fatty acid-binding protein hFABP, pyruvate decarboxylase, cytochrome-C oxidase, and succinate dehydrogenase. α1-Adrenoceptor stimulation of cardiomyocytes causes synthesis of angiotensin-1 that acts as an endothelin-releasing factor on smooth muscle cells in coronary arterioles, as it provokes endothelin release via NADPH oxidase and subsequent vasoconstriction [1011]. The ET B receptor impedes T-cell adhesion to endothelium, especially in cancers, characterizd by an overexpression of endothelial ET B receptor. 129 The ET A receptor 129. Endothelin-1 is produced at high concentrations by ovarian cancer cells. Endothelin-1 inhibits in vitro T-lymphocyte adhesion to endothelial cells, hence extravasation into the elicits T-cell homing on endothelium of lung vessels during allergy and inflammation. Increased release of NO mediates effects of ET B receptors with reduced expression of ICAM1 intercellular adhesion molecule. The sympathetic compartment of the autonomic nervous system is composed of preganglionic sympathetic neurons that have synapses with cell bodies of postganglionic sympathetic neurons in para-and prevertebral sympathetic ganglia. Postganglionic sympathetic neurons innervate smooth muscle layers of blood vessels and intestine, myocardium and nodal tissue, exocrine and endocrine glands and ducts, etc. During development, sympathetic neurons extend axons along paths, often jointly with artery trajectories, to innervate target tissues. Endothelin-3 released from the neural crest-derived smooth muscle layer of the external carotid arteries acts via ET A to direct extension of axons of a subset of sympathetic neurons from the superior cervical ganglion (located near the bifurcation of the common carotid artery) along the external carotid artery, which serves with its branches as gateway to appropriate organs [1013] . On the other hand, ET3-ET A signaling does not participate in projections to the internal carotid artery. Other factors expressed by the mesodermderived smooth muscle layer of the internal carotid artery are used for navigating projections from the superior cervical ganglion toward the internal carotid artery and its branches. Estrogens signal via 2 cytoplasmic and 1 plasmalemmal receptor. Nuclear receptors include estrogen receptor-α and -β (NR3a1-NR3a2; Sect. 6.3.5.1). Estrogen receptors ERα and ERβ are able to associate with plasmalemmal components. On the other hand, estrogens bind with high affinity to G-protein-coupled estrogen receptor (GPER or GPR30). Liganded G-protein-coupled receptor GPR30 promotes rapid estrogen signaling. G-protein-coupled estrogen receptor targets cAMP signaling, as it couples to Gs and Gi/o subunits of guanine nucleotide-binding proteins [5] . Plasmalemmal GPER not only regulates adenylate cyclase, but also primes extracellular release of tumor. The ET B receptor on the wetted surface of tumor endothelium prevents transendothelial migration from the blood stream of T lymphocytes, which then cannot infiltrate tumors [1012] . Suppression of ET B activity is required for tumor immunotherapy by increasing homing of tumor-specific T lymphocytes. proheparin-binding EGF-like growth factor (Sect. 8.2.5.2) . 130 In addition, G protein subunits, such as Gα i and Gβγ, interact with ERα. 131 The GPER receptor activates phosphatidylinositol 3-kinase and protein kinase-B, as well as members of the mitogen-activated protein kinase modules, such as extracellular signal-regulated protein kinases ERK1 and ERK2. The activation of the PI3K pathway by GPER occurs via GPER-mediated transactivation of the epidermal growth factor receptor (Sect. 7.7). In arterial smooth muscle cells, activated GPER increases intracellular calcium concentration. In cardiomyocytes, GPER activation ensures cardioprotection during myocardial ischemia-reperfusion events, as it reduces postischemic dysfunction and infarct size via the PI3K-PKB and ERK pathways [1015] . Agent PI3K is required, but not MAPK kinases that phosphorylate (activate) extracellular signal-regulated protein kinases. The GPER receptor inhibits mitochondrial permeability transition pore opening via the ERK pathway, hence protecting the heart against ischemiareperfusion injury [1016]. Fed mammals use glucose as the main metabolic fuel. However, short-chain fatty acids produced by the fermentation of dietary fibers by the intestinal colonic bacterial flora 132 also contribute to a significant proportion of daily energy requirement. Under starvation and ketogenic conditions, ketone bodies produced in the liver from fatty acids are used as the main energy sources. Feeding and fasting regulate chemical energy availability via the sympathetic nervous system. Feeding enables con-130. Heparin-binding epidermal growth factor-like growth factor (HBEGF) of the EGF family is synthesized as a type-1 transmembrane precursor (proHBEGF). At the cell surface, pro-HBEGF is a juxtacrine growth factor that signals to neighboring cells via an intercellular contact. The ectodomain of proHBEGF is shed and can serve as an N-terminal soluble ligand of EGFR ( N HBEGF or simply HBEGF) that can indirectly transactivate HER2, HER3, and HER4 heterodimerized with EGFR (HER1). 131. Plasmalemmal estrogen receptor-α is a palmitoylated protein. 17β-Estradiol reduces ERα palmitoylation and its interaction with caveolin-1 in a time-and dose-dependent manner [1014] . Palmitoylation of ERα enables rapid E 2 signaling via the ERK and PI3K-PKB pathways. Upon 17β-estradiol binding, ERα undergoes depalmitoylation and dissociates from caveolin-1 (inactivation-activation cycle). 132. Commensal bacterial communities are closely associated with the human skin, oral cavity, gastrointestinal tract, and the female genital tract. Gut flora consists of microorganisms (bacteria, fungi, and protozoa). Most bacteria belong to the species Bacteroides, Clostridium, Fusobacterium, Eubacterium, Ruminococcus, Peptococcus, Peptostreptococcus, and Bifidobacterium, as well as, but to a lesser extent, Escherichia and Lactobacillus. The major species are Bacteroides fragilis, Bifidobacterium adoloscentis, and Eubacterium areofaciens [1017] . The remainder is mainly Escherichia coli, Streptococcus viridans, Streptococcus salivaris, and lactobacilli. Anaerobic species outnumber aerobes (at least 10-fold). The Bacteroides-Prevotella group (Gram− anaerobes) and Clostridium species (Gram+ anaerobes) predominate [1018] . Fungi of the gut flora pertain to the genera Candida, Saccharomyces, Aspergillus, and Penicillium. Table 7 .60. Free fatty acid receptors and their main G-protein subunit transducers (Sources: [5, 1019] ; O3FAR: omega-3 fatty acid receptor). Long-chain saturated and unsaturated fatty acids (lcFFA) activate free fatty acid FFA 1 receptor, whereas short-chain fatty acids (scFFA) activate FFA 2 and FFA 3 receptors. The GPR84 receptor is stimulated by medium-chain free fatty acids (mcFFA). The GPR120 receptor mediates the anti-inflammatory and insulin-sensitizing effects of omega-3 fatty acids. Four genes -Ffar1 to Ffar3, and Gpr42 -form a cluster on human chromosome 19q13. Other sumption of chemical energy into thermal energy. During fasting, chemical energy is saved by the reduction of the sympathetic activity. Free fatty acids (FFA) serve not only as nutrients, but also as signaling molecules. Free fatty acid receptors (FFAR) are G-protein-coupled receptors. Four subtypes encoded by different genes exist: FFAR1, or GPR40, FFAR2, or GPR43, FFAR3, or GPR41, and GPR42 (Table 7 .60). Receptors GPR84 and GPR120 can also be activated by FFAs. Receptors FFAR1 and GPR120 are activated by medium-and longchain free fatty acids [1019] . 133 The GPR84 receptor is stimulated by medium-, but not long-chain, free fatty acids. Both FFAR2 and FFAR3 are activated by short-chain free fatty acids. The FFAR1 receptor is mainly expressed in pancreatic β cells, where it fosters insulin secretion [1020] . Receptors FFA 2 and FFA 3 are synthesized in adipocytes; FFA 3 also in enteroendocrine cells. The GPR84 receptor is manufactured in the spleen. The GPR120 receptor abounds in the intestine, where it promotes the secretion of glucagon-like peptide-1. Mid-and long-chain saturated and unsaturated fatty acids are ligands for FFAR1 receptor. The latter is coupled to the formation of inositol (1,4,5)-trisphosphate, Ca ++ influx, and activation of extracellular signal-regulated kinases ERK1 and ERK2. Linolenic acid targets FFA 1 on rat pancreatic β cells and reduces the voltagegated K + current via the cAMP-PKA axis to enhance β-cell excitability and insulin secretion [1019]. In mice, FFAR1 is also expressed in endocrine cells of the gastrointestinal tract, such as those that synthesize the incretin hormones glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide (GIP) [1019] . It is also observed on splenocytes and human peripheral blood mononuclear cells. The FFAR2 receptor is synthesized predominantly in immunocytes. Its expression is also induced during the differentiation of leukocyte progenitors into monocytes or neutrophils. It can be found in enteroendocrine cells that produce peptide-YY and mucosal mastocytes. The FFAR2 receptor abounds in adipocytes (but not FFAR3), where it may be implicated in the production of leptin, a potent anorexigenic hormone [1019]. Short-chain fatty acids such as propionate and ketone bodies control the amount of chemical energy, as they regulate the activity of sympathetic ganglia via the Gi/o-coupled receptor FFAR3 that triggers the Gβγ-PLCβ-MAPK pathway [1020]. A major short-chain fatty acid, propionate, supports the GPR41-mediated activation of the sympathetic nervous system. On the other hand, a ketone body -βhydroxybutyrate -produced during starvation or diabetes antagonizes FFAR3 receptor. Among medium-chain FFAs, capric, undecanoic, and lauric acids are the most potent agonists of the GPR84 receptor. In humans, GPR84 is expressed in the brain, heart, muscle, lung, kidney, liver, intestine, colon, thymus, spleen, and placenta, as well as leucocytes. In activated T lymphocytes, GPR84 participates in the regulation of early production of interleukin-4 [1019]. The GPR120 receptor, first identified as an orphan G-protein-coupled receptor, is bound by mid-and long-chain, saturated and unsaturated fatty acids, It is highly expressed in the human intestinal tract. Endocrine cells that produce glucagon-like peptide GLP1 in the large intestine synthesize GPR120. In addition, K cells that release glucose-dependent insulinotropic polypeptide in the duodenal and jejunal epithelia strongly express FFAR1, GPR119, and GPR120 [1019]. In mice, enteroendocrine STC1 cells that secrete GLP1 and cholecystokinin upon stimulation by FFAs are endowed with GPR120. The GPR120 receptor is also synthesized in adipocytes, type-2 taste cells of rat taste buds and Clara cells in lungs [1019]. In adipose tissue, the expression of GPR120 is higher in adipocytes than in stromal-vascular cells. Frizzled receptors (Fz; Sect. 10.3) that are activated by Wnt ligands trigger β-catenin-dependent (canonical) and -independent (non-canonical) signaling. The Wnt-Fz axis can raise intracellular calcium concentration, signal via Disheveled phosphoproteins to monomeric small GTPase Rac1 and Jun N-terminal kinase, as well as Rho and RoCK kinases, activate cGMP-specific phosphodiesterase PDE6, and heighten cAMP via heterotrimeric G proteins. Like other GPCRs, members of the Frizzled family (Table 7 .61) depend on β-arrestin scaffold protein for internalization and signaling. The Wnt-Fz signaling is controlled by: (1) additional ligands that can enhance (extracellular Norrin or R-spondins-1 to -4) or inhibit Fz (secreted Frizzled-related protein-1 to -5, extracellular Wnt inhibitory factor WIF1, and Dickkopf-1) as well as (2) modulatory proteins with positive (RYK and ROR1 and ROR2 kinases) and negative (extracellular Kremen-1 and -2) regulatory effects [5]. γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the central nervous system. Ionotropic γ-aminobutyric acid receptors (GABA A ; Sect. 2.5.3) are pentameric, ligand-gated, Cl − channels. Metabotropic γ-aminobutyric acid receptors (GABA B : Table 7 .64) are expressed in almost all neurons of the brain. They regulate synaptic transmission and signal propagation. They indeed control the activity of voltage-gated calcium (Ca V ) and inward rectifier potassium channels (K IR ). The GABA B receptor can indeed associate with Ca V 2.1 and Ca V 2.2 and K IR 3 channels [5] . Its main transducer is a subunit of the Gi/o family. Selective agonists of GABA B include 3-aminopropyl P methylphosphinic acid (3APMPA) and 3aminopropylphosphinic acid (3APPA). Metabotropic GABA B receptor is a heterodimer formed by 2 similar subunits GABA B1 and GABA B2 . The GABA B1 subunit alone binds cognate ligands (antagonists and agonists), but with much lower affinity (10-100 fold less) [5] . Calcium ion binds to GABA B1 to act as a positive modulator. Coexpression of GABA B1 and GABA B2 enables the transport of GABA B1 to the plasma membrane and generates a functional receptor. 134 The GABA B2 subunit mediates G-protein-coupled signaling. The GABA B2 subunit increases GABA B1 affinity for agonists [5] . Reciprocally, GABA B1 facilitates coupling of GABA B2 to G proteins. Therefore, the GABA B2 subunit does not signal by itself in response to γ-aminobutyric acid, but is essential for cell surface expression of the binding GABA B1 subunit and for signaling of the heterodimer, as GABA B1 transactivates GABA B2 . Several GABA B1 isoforms exist. The GABA B1a and GABA B1b isoforms are predominant subtypes in neonatal and adult central nervous system, respectively [5] . 135 In the central nervous system, the GABA B receptor is constituted by not only GABA B1 and GABA B2 , but also members of a subfamily of auxiliary K + channel tetramerization domain-containing proteins (KCTD) [1021] . These KCTD proteins increase the potency of agonists and accelerate receptor-effector coupling, hence G-protein signaling. Galanin receptors (Gal 1 -Gal 3 or GalR1-GalR3) are activated by galanin and galanin-like peptide (Table 7 .62). They localize to the peripheral and central nervous systems and endocrine system. The galanin family includes galanin and galanin message-associated peptide (GMAP) that are generated from the same peptide precursor on the one hand, and galanin-like peptide (GalP) encoded by a different gene and alarin, a GALP splice variant, on the other. Several neuropeptides are involved in the pathophysiology of depression, in addition to noradrenaline and serotonin. Galanin mainly inhibits both the noradrenergic and serotonergic networks via Gal 1 and Gal 3 receptors. It also impedes neurotransmission via Gal 2 and Gi/o subunits, but can stimulate neurotransmitter release via Gal 2 and Gq/11 subunits. Galanin hampers neurotransmission in memory acquisition, modulates food intake and sexual behavior, and participates in the control of pain. Galanin is also a neuroendocrine peptide of the gastrointestinal tract. Galanin precludes insulin release. 134. Subunit GABA B1 alone is not transported to the cell surface and remains non-functional. 135. The GABA B1a heterodimers on distal axons impede glutamate release in CA1-CA3 nerve terminals and GABA release onto the layer 5 pyramidal neurons. The GABA B1b heterodimers on dendritic spines mediate slow postsynaptic inhibition. Table 7 .63. Main G-protein subunit transducers and ligands of glucagon receptors (Source: [5] ; GHRH: growth hormone-releasing hormone; GIP: glucose-dependent insulinotropic polypeptide; GLP: glucagon-like peptide). ). Glutamate ionotropic receptors include members of N methyl D aspartate (NMDA; GluN1, GluN2a-GluN2d, and GluN3a-GluN3b) and α-amino 3-hydroxy 5-methyl 4-isoxasole propionic acid (AMPA; GluR1-GluR4), and kainate (GluK1-GluK5) receptor families. The GABA A receptors are ligand-gated ion channels. The GABA B receptors can couple to Ca V 2.1 and Ca V 2.2 as well as K IR 3 channels. Main transducer Glutamate metabotropic receptors mGlu 1 / 5 Gq/11 mGlu 2 -4 , mGlu 6 -8 Gi/o γ-Aminobutyric acid receptors GABA B Gi/o Ionotropic glutamate receptors include members of NMDA-and AMPA-type glutamate receptor channel family 139 that have a high relative permeability to calcium ions and are blocked by magnesium at rest potential. Hydrogen and zinc ions also inhibit NMDA receptor channel. Metabotropic glutamate receptors (mGlu 1 -mGlu 8 or mGluR1-mGluR8) are (Table 7.64) activated by L glutamate as well as L aspartate, L serine O P , 140 N acetylaspartylglutamate, and L cysteine sulfonic acid. Three groups of receptors have been defined according to their sequence, Gprotein coupling, and ligands: group 1 (mGlu 1 and mGlu 5 ), 2 (mGlu 2 and mGlu 3 ), and 3 (mGlu 4 and mGlu 6 to mGlu 8 ). Positive (potentiators) and negative allosteric modulators operate in the presence of agonists. L glutamate is the main neurotransmitter in the central nervous system that mediates fast excitatory neurotransmission by activating ionotropic glutamate receptors. Metabotropic glutamate receptors that are not located in the synaptic cleft prime slower effects. Type-1 mGluRs that operate in synaptic transmission are associated with slow synaptic excitatory currents and long-term remodeling. Kinetics of mGluR activation and deactivation determine features of synaptic transmission. Metabotropic glutamate receptors can be characterized by fast activation and slow deactivation. Activation time course can actually reach a minimum of approximately 10 ms, whereas deactivation time course nearly equals to 50 ms [1022] . Sensitization can develop over about 400 ms. Neuronal stimulation of astrocytic metabotropic glutamate receptors induces an astrocytic calcium wave that propagates to astrocytic endfeet close to brain arterioles. 139. NMDA: N methyl D aspartate; AMPA: α-amino 3-hydroxy 5-methyl 4-isoxasole propionic acid. 140. This L serine precursor ( L SOP) in the serine synthesis pathway is produced by phosphoserine (Ser P ) aminotransferase (PSAT) and metabolized to L serine by phosphoserine phosphatase (PSP). It is an agonist at group-3 metabotropic glutamate receptors. Active neurons then rapidly (latency < 2 s) trigger dilation of intracerebral arterioles. Astrocytic calcium signaling activates astrocytic large-conductance, Ca ++ -sensitive K + channels (BK) for local release of potassium into the perivascular space. A modest rise in extracellular potassium activates inward rectifier K + channels (K IR 2.1) of brain arteriolar smooth muscle cells, inducing membrane potential hyperpolarization and relaxation [1023] . 141 Augmented neuronal activity thus increases the local cerebral blood flow to supply sufficient amounts of glucose and oxygen (functional hyperemia). Homer proteins associate with metabotropic glutamate receptors mGluR1 and mGluR5 as well as inositol trisphosphate receptors to increase mGluR-stimulated Ca ++ signaling. In addition, Homer proteins facilitate anchoring and clustering of mGluR1 and mGluR5 in postsynaptic dendritic spines. Scaffolds of the Shank family of SH3-and multiple ankyrin repeat domain-containing proteins interact with both Homers and mGluRs to strengthen mGluR anchoring [835]. Glycoprotein hormone receptors are activated by heterodimeric glycoproteins made up of a common αand a β chain that confers the specificity of these glycoproteic heterodimers. These glycoprotein hormones include: (1) follicle-stimulating hormone (FSH, or follitropin); (2) luteinizing hormone (LH, a.k.a. lutropin and, in males, interstitial cell-stimulating hormone [ICSH]); (3) choriogonadotropin (or chorionic gonadotropin); and (4) thyroid-stimulating hormone (TSH, or thyrotropin; Table 7 .65). Follicle-stimulating hormone receptor (FSHR) in gonads is targeted by pituitary FSH glycoprotein for gonadal development, maturation at puberty, and sustained gamete production during the reproductive phase of life. Follicle-stimulating and luteinizing hormones that are both synthesized and secreted by the anterior pituitary gland act synergistically in reproduction. At the moment of menstruation, FSH initiates follicular growth. Stimulated FSHR not only prevents atresia of early antral follicles, but also promotes granulosa cell proliferation, estrogen synthesis, and LHR expression [1024] . An acute release of LH (LH surge) triggers ovulation and corpus 141. Elevation of the concentration of external K + that depolarizes vascular smooth muscle cells induces vasoconstriction of cerebral arteries and arterioles. Main transducer Potency order GnRH 1 Gq/11 GnRH1 > GnRH2 GnRH 2 Gq/11 GnRH2 > GnRH1 luteum development. In the ovary, the number of FSHRs increases during follicular maturation under low levels of FSH and decays after ovulatory LH surge. In both males and females, FSH stimulates the maturation of germ cells. In males, it stimulates Leydig cell production of testosterone, enables persistance of Sertoli cells during gonadal development, and causes inhibin secretion in these cells. Unlike LHR, gonadal FSHR expression is specific to cell type [1024] . In the ovary and testis, FSHRs are expressed on granulosa cells of developing follicles and Sertoli cells, respectively. Follicle stimulating hormone receptor interacts with arrestins and G-proteincoupled receptor kinases [1024] . Immature FSHR also associates with chaperone proteins calnexin, calreticulin, and disulfide isomerase as well as 14-3-3 adaptor protein, APPL (adaptor protein, phosphoTyr interaction, pleckstrin homology domainand leucine zipper-containing protein), and FOXO1a. Signaling mediated by the thyroid-stimulating hormone receptor and the Gprotein subunit Gs can be enhanced by receptor endocytosis [11] . Gonadotropin-releasing hormone (GnRH) is a hypothalamic decapeptide (Vol. 2 -Chap. 1. Remote Control Cells -Sect. Endocrine System and Hormones). Isoforms GnRH1 and GnRH2 as well as their cognate receptors GnRH 1 and GnRH 2 exist in mammals (Table 7 .66). The GnRH 1 receptor is produced primarily in pituitary gonadotrophs and contribute to the central control of reproduction. Receptors GnRH 1 and GnRH 2 couple primarily to Gq/11, but also to Gs and Gi subunits. The GnRH 2 receptor can also target protein kinases [5]. Histamine 142 is a neurotransmitter of the nervous system 143 and a signaling molecule in the immune system, skin, and gut. 144 Endothelial cells do not synthesize histamine and histamine receptors, but they can take up histamine. Histamine binds to 4 known metabotropic histamine receptors (H 1 -H 4 or H1R-H4R). These receptor subtypes associate with different types of G proteins to prime various effects (Table 7 .67). Receptor H 3 abounds in the brain; H 4 receptor lodges mainly in peripheral tissues. Receptors H 1 and H 2 are mostly excitatory; H 3 homoand heterodimers are inhibitory [1025] . Bordetella pertussis that can cause encephalomyelitis and disrupts the bloodbrain barrier. On the other hand, H 1 on endothelial cells counteracts Bordetella pertussis effect. It actually reduces the permeability of the blood-brain barrier [1029] . Histamine mediates the stress-induced surges of adrenocorticotropic hormone, β-endorphin, and vasopressin from the pituitary gland (hypophysis) [1025]. It controls stress-related activity of aminergic networks (serotonin-, noradrenaline-, dopamine-, and acetylcholine-containing neurons). The preferential site of histamine-induced suppression of food intake is Histamine stimulates neurons in the supraoptic nucleus that release the antidiuretic hor-142. I.e., amine in biological tissues. This organic nitrogen compound s also called imidazolethylamine or imidazolethanamine. Histamine is synthesized from histidine taken up into the cerebrospinal fluid and neurons by amino acid transporter via oxidative decarboxylation by histidine decarboxylase. It is stored and transported into a vesicle using the vesicular monoamine transporter VMAT2, and, upon appropriate cues, released and methylated (inactivated) by neuronal histamine N methyltransferase using S adenosyl methionine. The latter enzyme also resides in blood vessel walls and processes circulating and mastocyte-released histamine. The main histamine-degrading enzyme in peripheral organs is diamine oxidase that converts histamine into imidazoleacetic acid [1025] . 143. In the mammalian brain, histaminergic neurons localize exclusively in the tuberomamillary nucleus of the posterior hypothalamus and send their axons to the entire central nervous system, like other amine networks [1025] . All amine networks rely on autoreceptors that yield a negative feedback on excitability and amine neurotransmitter release and synthesis. Histaminergic neurons are active only during waking; they maintain wakefulness and attention. Mutual interactions with other transmitter networks are involved in higher brain functions (sleep-wake regulation, circadian and feeding rhythms, immunity, learning, and memory. Although histamine is the main neurotransmitter in the tuberomamillary nucleus, cotransmitters include GABA, galanin, enkephalins, thyrotropin-releasing hormone, and substance-P [1025]. Glutamatergic fibers from the cortex and hypothalamus secrete glutamate that excites neurons of the tuberomamillary nucleus. On the other hand, glycine inhibits a subpopulation of histaminergic neurons. In addition, gabaergic cues from mostly hypothalamic origin suppress the firing of histaminergic neurons. Aminergic (noradrenergic via gabaregic and serotoninregic) and cholinergic neurons also send projections to histaminergic neurons. Nucleotides excite histaminergic neurons using ionotropic and metabotropic receptors. 144. In neuroepithelial and hematopoietic cells, histamine participates in contraction of airway and intestinal smooth muscles, but vasodilation, as well as gastric acid secretion, mastocyte-based innate and acquired immunity, allergy, and inflammation, T-cell-associated immunity modulation, and control of epi-and endothelial barriers [1025]. likely the satiety center of the ventromedial hypothalamus. Histamine effects on food intake are associated with other facors, such as neuropeptide-Y, peptide-YY, and bombesin. In addition, orexigenic action of orexins and anorexigenic effects of leptin and glucagon-like peptide-1, which depend on corticotropin-releasing hormone released by neurons of the paraventricular nucleus rely on H 1 receptor [1025] . The brain histamine network controls thermogenesis, mostly via feeding and motor activity [1025] . The body's autonomic response that regulates heat conservation and production relies on the paraventricular nucleus and dorsomedial hypothalamus, and the nucleus raphe pallidus, respectively. Most of the structures implicated in thermoregulation are targets of histaminergic innervation. Histaminergic activity exhibits a circadian rhythm with high levels during the day and low levels during the night [1025] . Histamine can entrain molecular clockworks outside the suprachiasmatic nucleus. The Gq/11-coupled H 1 receptor activates phospholipase-C, thereby supporting IP 3 -dependent release of Ca ++ from its intracellular stores and diacylglycerolprimed activation of protein kinase-C. The latter forsters Ca ++ influx through voltage-dependent calcium channels, transient receptor potential channels of the TRPC family, and Na + -Ca ++ exchanger. Other effects include production of arachidonic acid, nitric oxide, and cGMP using Gi/o-mediated activation of phospholipase-A2, Ca ++ -dependent NOS, and NO-dependent guanylate cyclases, respectively. Moreover, H 1 activate AMPK kinase and nuclear factor-κB [1025] . Glia cells produce H 1 and H 2 receptors. Histamine promotes release of neurotrophins and cytokines from astrocytesas well as ATP in the hypothalamus [1025] . Histamine causes the opening of the blood-brain barrier, mainly via H 2 receptor. Gs-coupled H 2 receptor stimulates adenylate cyclase, thereby elevating intracellular cAMP concentration, then activating protein kinase-A and CREB transcription factor [1025] . In addition, cAMP can interact with hyperpolarization-activated cation HCN2 channel. Upon PKA-mediated phosphorylation, H 2 represses Ca ++ -activated, small conductance K + channel (SK), which enables the long-lasting (seconds) afterhyperpolarization after action potentials in pyramidal cells, as well as K V 3.2 channel in interneurons involved in fast spiking [1025] . Gi/o-coupled H 3 receptor links to Ca V 2.1 and Ca V 2.2 channels [1025] . Due to crosstalk with other GPCRs, H 3 can initiate Gq/11 signaling and activate phospholipase-A2, the PKB-GSK3 axis, and MAPK module to control axonal and synaptic remodeling. Gi/o-coupled H 4 receptor is expressed mainly in peripheral cells of blood, lung, liver, spleen, and gut [1025] . It may also reside in some regions of the brain. Endogenous peptides with an arginine-phenylalanine-amide motif: (1) chemotaxis-inhibitory kisspeptin-54 (KP54 or metastin) cleaved from its precursor, a Kiss1 gene product; (2) pain-modulatory neuropeptide-FF (NPff); (3) prolactin-releasing peptide (PRP); and (4) pyroglutamylated arginine-phenylalanine (QRF)-amide peptide (QRFP) bind to their cognate receptors [5] (Table 7 .68). Latrophilin receptors (Lphn1-Lphn3; a.k.a. calcium-independent receptors for latrotoxin [CIRL 1 -CIRL 3 ]) constitute a group of GPCRs of the class-B secretin family [835] . They operate in both cell adhesion and signal transduction. They are located in pre-and postsynaptical membranes in the central nervous system. They connect to postsynaptic proteins, such as SH3-and multiple ankyrin repeat domaincontaining proteins Shank1 to Shank3, a.k.a. somatostatin receptor-interacting protein (SstRIP) and proline-rich synapse-associated proteins ProSAP1 and ProSAP2, respectively [835]. Leukotrienes comprise 5-lipoxygenase-derived family of short-lived eicosanoid lipid mediators that modulate vascular function and inflammatory cell reactivity. Leukotrienes as well as lipoxins are active metabolites derived from arachidonic acid. They are mainly involved in auto-and paracrine signaling. 5-Lipoxygenase (5LOx) in conjunction with arachidonate 5-lipoxygenase-activang protein (ALOx5AP), 145 an integral membrane protein of the nucleus membrane, converts arachidonic acid into 5-hydroperoxy eicosatetraenoic acid (5HPETE) that spontaneously reduces to 5-hydroxy eicosatetraenoic acid (5HETE). 5-Lipoxygenase converts 5HETE into leukotriene-A4, an unstable epoxide. In cells equipped with LTa4 hydrolase, such as neutrophils and monocytes, LTa4 is converted to leukotriene-B4. In cells that possess LTc4 synthase, such as mastocytes and eosinophils, LTa4 gives rise to leukotriene-C4. Hydrolysis of LTc4 produces leukotriene-D4 and -E4. Leukotrienes include cysteinyl-leukotrienes (cysLTs: LTc4, LTd4, and LTe4) and dihydroxy-leukotriene (LTb4). Cysteinyl leukotrienes secreted by basophils and mastocytes together constitute the slow-reacting substance of anaphylaxis, as they trigger a prolonged, slow contraction of smooth muscle cells, especially vasoconstriction and bronchoconstriction. Leukotriene receptors are activated by leukotrienes LTb4, LTc4, LTd4, LTe4, 12 S HETE, and 12 R HETE (Table 7 .69). In addition to their receptors and ALX 145. A.k.a. five-LOx-activation protein (FLAP). Table 7 .69. Leukotriene receptors, their main targeted G proteins, and order of ligand potency (Sources: [5, 736] ; EPA: eicosapentaenoic acid; fMLP: formyl methionyl-leucylphenylalanine; HETE: hydroxyeicosatetraenoic acid; HPETE: hydroperoxyeicosatetraenoic acid; 5oxoETE: 5-oxo-eicosatetraenoic acid; RARRes2CTP [ChCTP]: retinoic acid receptor [RAR] responder-2 C-terminal peptide [chemerin C-terminal peptide; chemoattractant chemerin is also called tazarotene-induced gene-2 protein (TIG2)]; RvE1: resolvin-E1 [EPAderived (in the presence of aspirin), non-classic eicosanoid; inflammation reducer]). Receptors CysLT 1 and CysLT 2 are coexpressed by most myeloid cells. The CysLT 2 receptor suppresses CysLT 1 expression. The RS nomenclature (Latin rectus [R]: right behavior, straight, in a straight line, upright, direct path [used in the sense of dexter: right, on the right side]; sinister [S]: left, at the left side) for an enantiomer (Greek ναντιoς: opposite) defines the chirality (Greek χ ιρ: strong-handed) of a subtype of amino acid or carbohydrate (X) among 2 stereoisomers ( R X and S X). Main transducer Potency order RvE1 > RARRes2CTP >18 R HEPE > EPA lipoxin receptor, leukotrienes connect to enzymes of their metabolism (glutathione S-transferase-2, or LTc4 synthase, γ-glutamyl transpeptidase, and several aminopeptidases) and peroxisome proliferator-activated PPARα (nuclear receptor NR1c1). Lipoxins are short-lived, endogenous, non-classical eicosanoids generated by lipoxygenases that act as anti-inflammatory mediators. Leukotriene-B4 and related hydroxyacids target BLT receptors BLT 1 and BLT 2 . Cysteinyl-leukotrienes activate CysLT receptors CysLT 1 and CysLT 2 , as well as LTe4-specific CysLT E receptor. These leukotriene GPCRs are classified as either chemoattractants (BLT 1 and BLT 2 ) or nucleotide receptors (CysLT 1 and CysLT 2 ). Both BLT and CysLT receptors via Gq and Gi elicit a large increase in intracellular calcium concentration and a decrease in cAMP level, as well as kinase activation (i.e., extracellular signal-regulated protein kinases ERK1 and ERK2) to promote cell differentiation and chemotaxis. Main effects of leukotrienes and lipoxins are given in Table 7 .70. Lipoxins LXA4 and LXB4 146 activate lipoxin receptor ALX, a.k.a. formyl peptide receptor FPR2, formyl peptide receptor-like protein FPRL1, and formyl peptide 146. Lipoxins are eicosanoids that are generated within the vascular lumen by leukocytes. Lipoxins-A4 and -B4, as well as some peptides, are high-affinity ligands for LXA4 receptor (LXA4R), also called formyl peptide receptor-like receptor FPRL1. Similarly to leukotrienes, LXA4 can form cysteinyl-lipoxins LXC4, LXD4, and LXE4. At subnanomolar concentra- . Neuroprotective peptide non-formylated humanin (as well as much more potent N-formyl humanin) tethers ALX and prevents Aβ(1-42) activity [1030] . Peptide PACAP27 that also activates G-protein-coupled vasoactive intestinal peptide receptor VIPR 1 (VPAC 1 ) stimulates α M -integrin (CD11b) in human neutrophils [1030] . Vasoactive intestinal peptide also links to Gs-coupled vasoactive intestinal peptide receptors VIPR 1 and VIPR 2 (VPAC 2 ) to provoke smooth muscle relaxation, exocrine and endocrine secretion, and water and ion flux in lung and intestinal epithelia. It stimulates ALX to activate phosphatidylinositol 3-kinase and extracellular signal-regulated kinase and cause α M -integrin synthesis [1030]. ). Signaling from LTb4 in granulocytes is mediated by Gi as granulocytes abundantly express Gi, mainly Gi2, whereas nervous cells mainly produce Gi1 and Go. LTb4-induced calcium mobilization results from Gq activation. BLT1-mediated phospholipase-C activation is mediated by Gi6 and Gβγ. BLT2 is less efficient for calcium mobilization than BLT1. BLT2 induces chemotaxis via Gi and the Rac-ERK-ROS cascade. LTd4 activates CysLT1 to produce second messengers diacylglycerol, inositol 3-phosphate, and Ca ++ . Subsequently, protein kinase-C and phospholipase-A2 are activated, the latter generating arachidonic acid. LTd4 primes constriction of smooth muscle cells in bronchi and intestine via protein kinase-C . Smooth muscle cells (lung, intestine), blood leukocytes CysLT 2 Heart, spleen, adrenal medulla, brain Blood leukocytes ALX Lung, spleen Blood leukocytes ALX is involved in serum amyloid-A-stimulated interleukin-8 production and nuclear factor-κB activation. Although ALX activity regulation is similar to that of FPR1, FPR2/ALX differs from FPR1 receptor. Gelsolin-derived peptide PBP10 selectively blocks ALX-mediated mobilization of granules and production of oxygen radicals [1030] . Oxoeicosanoid receptor OxER1 (or GPR170) is activated by endogenous chemotactic eicosanoids oxidized at the C5 position [5] . 5-oxo-Eicosatetraenoic acid (5ox-oETE) is its most potent agonist. Resolvin receptors are activated by anti-inflammatory resolvin-E1 (RvE1) that derives from ω-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) by aspirin-modified cyclooxygenase and lipoxygenase [5] . Leukotrienes are synthesized not only by multiple cell types, but also via transcellular metabolism involving different cells, such as neutrophils, platelets, and vascular cells (Table 7 .71). Primary expression of leukotriene receptors is given in Table 7.72. Leukotriene-B4, a chemotactic and immune-modulating agent implicated in allergic and inflammatory reactions, acts via G-protein-coupled receptors BLT 1 and BLT 2 . Receptors BLT 1 and BLT 2 signal via 3 classes of G protein subunits, Gi, Gqlike, and Gz. 148 Leukotriene-B4 149 is a chemoattractant produced by neutrophils, macrophages, and mastocytes for polymorphonuclear leukocytes and T lymphocytes. Leukotrienes are involved in arteriosclerosis. In mature endothelial cells, LTb4-BLT signaling enables angiogenesis. Endothelial cell migration depends on CysLT receptors. Moreover, 12LOx-dependent pathway via 12HETE and BLT 2 is involved in VEGFa-induced angiogenesis. Embryoid bodies derived from human embryonic pluripotent cells differentiate into endothelial cells as well as different hematopoietic lineages (monocytes and macrophages, T lymphocytes, NK cells, and dendritic cells). Leukotrienes LTb4 and, to a lesser extent, LTd4 concomitantly with their cognate receptors BLT and CysLT are expressed in embryonic stem cells during embryogenesis, particularly endothelial plexus during the later phase of vasculogenesis and angiogenesis, at least in rodents. 150 Myeloid-like cells that synthesize 5LOx and its cofactor FLAP produce LTb4 that operates as a paracrine agent to stimulate endothelial progenitors via BLT receptors to initiate vasculogenesis [1034] . Leukotriene action in differentiating embryonic stem cells is mediated via extracellular signal-regulated kinase ERK1 and ERK2. Lysophospholipids are built on a glycerol backbone and are defined by the nature of the phosphate groups linked to the carbon atom at position 3 (stereospecific nomenclature sn3). They contain a single fatty acid side chain. On the other hand, phospholipids typically contain saturated and unsaturated fatty acids at the sn1 and sn2 positions, respectively. Lysophospholipids can be generated from phospholipids by phospholipases that cleave the sn1 or sn2 acyl fatty acids. The sn2 acyl side chain of lysophosphatidic acid (LPA) is unstable and moves to the sn1 position, generating the predominant form of LPA: 1-acyl 2-hydroxy sn-glycero 3-phosphate (Table 7 .73). The sequential actions of phospholipases PLD and PLA2 can generate lysophosphatidic acid from phosphatidic acid (PA). Structural and functional diversity arises from differences in fatty acid moieties esterified to sn1 that may influence LPA binding to its receptors. The synthesis of LPA involves either the sequential action of secretory phospholipase-A and lysophospholipase-D, or phospholipase-D and cytosolic 148. The expression of BLT receptors can be enhanced in endothelial cells by lipopolysacharides, cytokines, such as tumor-necrosis factor-α and interleukin-1β, and LTb4 [1033]. 149. LTb4 Leukotriene is synthesized from arachidonic acid by the concerted action of LTa4 hydrolase and 5-lipoxygenase, which is assisted by 5-lipoxygenase-activating protein. 150. Myeloid-like, CD11b+ (α M -integrin) embryonic stem cells are also 5LOx+ and FLAP+, whereas endothelial-like, VEGFR2+, CD105+ (endoglin of the TGFβ receptor complex) embryonic stem cells are non-synthesizing leukotriene cells. Table 7 .73. Main axes of lysophosphatidic acid (LPA) synthesis, either from phosphatidic acid (PA) using phospholipases (PL), or from lysophosphatidylcholine (lysoPC) using autotaxin, or lysophospholipase-D (lysoPLD). Autotaxin is produced in high endothelial venules, where it can support transendothelial migration and lymphocyte entry into lymph nodes and secondary lymphoid organs, as well as lymphocyte migration into inflammatory sites. PC-LysoPC-LPA sPLA2 and LysoPLD, or PLD and cPLA2 PA-LPA PLD and PLA2 Table 7 .74. Endothelial differentiation gene (EDG) family of GPCRs. Gene Name Ligand EDG1 S1P 1 S1P EDG2 LPA 1 LPA EDG3 S1P 3 S1P EDG4 LPA 2 LPA EDG5 S1P 2 S1P EDG6 S1P 4 S1P EDG7 LPA 3 LPA EDG8 S1P 5 S1P PLA. The second major axis of LPA synthesis uses lysophospholipase-D (lysoPLD), or autotaxin, that cleaves the choline group from lysophosphatidylcholine. In addition, membrane-bound phosphatidic acid-selective phospholipase-1α (MPAPLA1), or lipase-H (LipH), is a 2-acylLPA-producer. The generation of LPA can also be achieved by phosphorylation of monoacylglycerol (MAG) by MAG kinases. The most prominent source of LPA in the blood circulation (at micromolar concentrations) arises from autotaxin. Lysophosphatidic acid circulates bound to serum proteins, especially albumin and gelsolin, a member of the extracellular actin scavenger pool that depolymerises and removes from the blood actin released in the extracellular medium to avoid thrombus creation. In response to inflammation, LPA diffuses into the extravascular space, where it supports cell chemotaxis and proliferation, cytokine and chemokine secretion, platelet aggregation, and smooth muscle cell contraction (vasoconstriction). Glycerol-and sphingosine-based phospholipids are abundant components of cellular membranes. They are metabolized into eicosanoids and lysophospholipids, such as lysophosphatidic acid, lysophosphatidylcholine, sphingosylphosphoryl choline, and sphingosine 1-phosphate. G-protein-coupled receptors for lysophospholipids include 12 identified members. The lysophospholipid GPCR family, i.e., the endothelial differentiation gene (EDG) family of GPCRs, encompasses receptors for sphingosine 1-phosphate and lysophosphatidic acid, as well as lysophosphatidylcholine and sphingosylphosphoryl choline psychosine groups [1035] (Table 7 .74). Upon receptor activation, plasmalemmal phospholipids are metabolized into lysophospholipid mediators, such as sphingosine 1-phosphate and lysophosphatidic acid. These messengers regulate angiogenesis, cardiac development, neuronal survival, and immunity [1035] . Lysophosphatidic acid (LPA), the simplest natural phospholipid, 151 is produced by: (1) hydrolysis of phosphatidic acid that localizes mainly at the inner leaflet of the plasma membrane by phospholipase-A1 or -A2 and (2) conversion of lysophospholipids, such as lysophosphatidylcholine (LPC), 152 lysophosphatidylethanolamine (LPE), and lysophosphatidylserine (LPS), which are also generated from membrane phospholipids by phospholipase-A1 or -A2, to LPA by lysophospholipase-D, or autotaxin. 153 This bioactive lipid mediator involved in stress fiber formation using the Rho-RoCK axis 154 and cell proliferation, differentiation, survival, and motility. Lysophosphatidic acid is dephosphorylated (inactived) by lipid phosphate phosphohydrolases (LPP) into monoacylglycerol. The LPP phosphatase has its catalytic site oriented toward the external face of the plama membrane. Lysophosphatidic acid can be synthesized outside the cell by membrane or secreted enzymes, such as secreted phospholipase-A2 (sPLA2) and lysophospholipase-D (sLysoPLD), especially after membrane lipid reorganization during inflammation. Extracellular LPA that serves as lipid mediator is water soluble. It resides in many biological fluids, where it is maintained in solution owing to its connection to 151. Lysophosphatidic acid is composed of a single fatty acyl chain, a glycerol backbone, and a phosphate group. In fact, many LPA species exist. These LPA species possess an alkyl or alkenyl ether-linked fatty acyl chain of various lengths and unsaturation degrees. A saturated or unsaturated fatty acid chain is esterified either at the sn1 (1-acylLPA; e.g., 1-palmitoylLPA) or sn2 (2-acylLPA; e.g., 2-arachidonoylLPA) position (sn: stereospecific nomenclature) of the glycerol backbone of LPA phospholipid. Ether-linked LPAs carry an alkyl-(alkylLPA; e.g., hexadecylLPA) or an alkenyl-linkage (alkenylLPA) at the sn1 position [1036] . Activity of LPA depends on: (1) the length and the unsaturation level of the carbon chain attached to the glycerol backbone and (2) the type of linkage and its position. Lysophosphatidic acid is indeed synthesized via numerous routes. In particular, autotaxin, a lysophospholipase-D, removes the choline group from lysophosphatidylcholine. 152. Lysophosphatidylcholine abounds in biological fluids linked to albumin or lipoproteins. It is synthesized from phosphatidylcholine by PLA2 or lecithin cholesterol acyltransferase (LCAT). 153. Autotaxin (Atx; a.k.a. lysophospholipase-D and type-2 ecto-nucleotide pyrophosphatase phosphodiesterase) is a transmembrane ecto-enzyme. It is upregulated by certain peptide growth factors. Soluble autotaxin derives from a membrane-bound form by proteolytic cleavage. 154. In 1960, Vogt observed that LPA causes the contraction of isolated rabbit duodenum. Afterward, the contraction of smooth muscle cells generated by several LPA species was demonstrated on vascular cell types. G-protein subunit transducers, and ligands (Sources: [5, 1036] ; EDG: endothelial differentiation gene product). Lysophosphatidic acid also binds PPARγ receptors. In addition to the 3 G-protein-coupled receptors of the endothelial differentiation gene (EDG) family (LPA 1 -LPA 3 ), lysophosphatidic acid operates via 3 other homologous receptors (LPA 4 -LPA 6 ) that are structurally distinct from the EDG family LPA receptors. The latter are involved, in particular, in angiogenesis and platelet activation. The LPA receptors of the EDG family that preferntially link to acylLPA than alkylLPA provoke intracellular Ca ++ influx, inositol phosphate production, adenylate cyclase inhibition, and mitogen-activated protein kinase activation. Receptors LPA 6 , or P2Y 5 , and LPA 8 , or P2Y 10 its carrier albumin. In addition to albumin, other proteins, such as fatty acid-binding proteins and gelsolin, link to LPA. Almost all cells, except in the liver, coexpress several types of LPA receptors (LPA 1 -LPA 6 ; Table 7 .75). Receptors LPA 4 and LPA 6 correspond to P2Y 9 and P2Y 5 , respectively [1037, 1038] . The P2Y 9 receptor is most closely related to P2Y 5 that is coupled to the G13-Rho pathway. It is relatively close to nucleotide receptors P2Y 1 , P2Y 4 , and P2Y 6 as well as lipid receptors of the group of proton-sensing GPCRs 155 and other lipid receptors, such as platelet-activating factor 156 receptor and leukotriene receptors CysLT 1 and CysLT 2 . Lysophosphatidic acid receptors are coupled to G-protein subunits Gs, Gq, Gi, and/or G12/13 (Tables 7.75 and 7.76) . Activated Gq stimulates phospholipase-C that activates protein kinase-C for calcium influx. Activated Gi targets 3 pathways, as it: (1) inhibits adenylate cyclase; (2) stimulates Ras-mitogen-activated protein ki-155. Lipid ligands modulate action of protons that activate proton-sensing G-protein-coupled receptors. Proton-sensing GPCRs include: (1) Gs-coupled GPR4 that is targeted by sphingosylphosphorylcholine and lysophosphatidylcholine; (2) psychosine receptor GPR65 (or T-cell death-associated gene product TDAG8) that activates adenylate cyclase; (3) GPR68 (or ovarian cancer GPCR OGR1); and (4) GPR132 (or G2A), a high-affinity receptor for lysophosphatidylcholine. Sphingosyl-phosphorylcholine, lysophosphatidylcholine and psychosine are high-and low-affinity ligands for GPR4 and GPR65, respectively. Lipids favor effects of protons as activators of GPR4. Psychosine and its related lysosphingolipids behave as antagonists of protein-sensing receptors [1039] . 156. Platelet-activating factor is a phospholipid (acetyl-glyceryl-ether phosphorylcholine) produced by neutrophils, basophils, platelets, and endothelial cells. nase cascade; and (3) activates phosphatidylinositol 3-kinase, guanosine nucleotideexchange factor TIAM1 (RacGEF), hence small GTPase Rac, as well as protein kinase-B. Hence, LPA has mitogenic and anti-apoptotic effects. Activated LPA 1 and LPA 2 coupled to G12/13 stimulates small GTPase RhoA that causes cytoskeletal contraction and favors cell motility and adhesion. Lysophosphatidic acid receptor LPA 2 interacts with scaffold membrane-associated guanylate kinase, WW and PDZ domain-containing protein MAGI3 to enhance receptor-mediated activation of extracellular signal-regulated kinase. On the other hand, LPA 2 connects to 2 other PDZ domain-containing scaffolds, RhoGEF11 and RhoGEF12 to potentiate LPAR2-induced stimulation of Rho signaling on cytoskeleton dynamics [835] . Therefore, LPA 2 can activate PLCβ, ERK, or Rho GTPase according to the synthesis level of PDZ domain-containing scaffold, hence cell type. Lysophosphatidic acid receptor LPA 2 (or LPAR2), like purinergic receptor P2Y 1 , parathyroid hormone receptor PTH1R (or PTH 1 ), and metabotropic glutamate receptor mGluR5 can link to Na + -H + exchanger regulatory factors NHERF1 and NHERF2 independently of agonists to enhance Protein NHERF2 can also bridge LPA 2 to cystic fibrosis transmembrane regulator Cl − channel. On the other hand, β2AR and KOR opioid receptors also connect to NHERF proteins, but in an agonist-dependent manner [835] . Due to its unique C-terminal binding domain, LPA 2 bind to several PDZ motifcontaining proteins (NHERFs, RhoGEF11, RhoGEF12, and membrane-associated guanylate kinase with an inverted domain structure protein MAGI3) [1040] . These scaffolds modulate LPA-induced activation of ERK and/or RhoA GTPase. Both NHERF2 and MAGI3 can recruit phosphatase and tensin homolog to repress the PI3K-PKB signaling. In addition, NHERF2 can also serve as a scaffold for 3phosphoinositide-dependent protein kinase PDK1 that activates AGC family kinases such as PKB. Therefore, NHERF2 restricts or promotes the PI3K-PKB pathway according to the relative synthesis rate of PTen and PDK1 enzymes. In addition to NHERF2 scaffold, LPA 2 interacts with thyroid hormone receptor interactor TRIP6 and apoptosis-inducing factor Siva-1. The LPA 2 -TRIP6-Siva-1 complex and NHERF2 enable LPA 2 -mediated protection against apoptosis [1040] . Whereas LPA 1 and LPA 2 can be coupled to Gi, thus inhibiting adenylate cyclase, LPA 3 can be tethered to Gs, thereby activating adenylate cyclase. Liganded LPA 4 coupled to Gs and Gq subunits increases intracellular concentrations of Ca ++ and cAMP messengers. Receptor LPA 4 coupled to G12/13 proteins activates Rho GTPase, thereby causing cytoskeleton remodeling. Therefore, LPA 4 can counteract the effect of LPA 1 , as Rho and Rac exert a mutual inhibition. Receptor LPA 5 can activate adenylate cyclase independently of Gα s [1036] . It also mobilizes Ca ++ ions. In mice, LPA 5 is highly produced in the small intestine and stomach as well as dorsal root ganglion. In mouse small intestine, LPA 5 is synthesized not only in epithelial cells, but also in intraepithelial lymphocytes. 157. Many NHERF-binding partners, such as TRPC channels, various isoforms of PLCβ, protein kinases PKC and PKD, are components of Gq-PLCβ pathways. Plasmalemmal protein LPA 5 is the most prevalent LPA receptor in human mastocytes [1042] . 158 Cell proliferation caused by LPA stimulation of mastocytes results from the activity of LPA 1 and LPA 3 , whereas cytokine generation is induced by LPA 2 receptor. Receptor LPA 5 is responsible for release of CCL4 chemoattractant launched by LPA [1042] . Lysophosphatidic acid is a potent pro-aggregating factor for platelets. In humans, platelets synthesize LPA 1 to LPA 6 transcript; the most abundant are LPA 4 and LPA 5 transcripts [1041] . At the protein level, among LPA receptors, platelets produce mainly LPA 5 receptor. In humans, platelets aggregate more potently in response to alkylLPA than to corresponding acylLPA [1036] . Lysophosphatidic acid induces not only smooth muscle contraction, platelet aggregation, and increase in endothelial permeability [1043] , 159 but also cell survival, proliferation, and migration, as well as attachment of myeloid cells to endothelium (Table 7 .76). In addition it causes gap-junction closure and tight-junction opening, favors wound healing, and elicits the production of endothelin, angiogenic factors (vascular endothelial growth factor and interleukin-6 and -8), urokinase-type plasminogen activator, 160 metallopeptidases such as MMP2 and ADAM17 [1044] . Lysophosphatidic acid is a ligand for the transcription factor perixosome proliferating activating receptor-γ. Five G12/13-coupled LPA receptors can contribute to angioand lymphangiogenesis. Transcription of LPA is activated by growth factors. Lysophosphatidic acid can indirectly regulate cell functions by stimulating G-protein-coupled receptorregulated transmembrane metallopeptidase that cleaves precursor heparin-binding EGF-like growth factor (HBEGF) at the plasma membrane and activating EGF receptor (Sect. 8.2.5.2). Signaling molecules expressed in nociceptive sensory neurons include vanilloid receptor VR1, purinergic receptors such as P2X 3 , and tetrodotoxin-insensitive voltage-gated sodium channels that convey most of inward flux during upstroke and shoulder during the falling phase of nociceptor action potentials, with significant contribution from tetrodotoxin-sensitive sodium channels and voltage-dependent calcium channels, respectively, and regulate duration of action potentials. Several GPCRs are expressed in nociceptive sensory neurons, such as bradykinin, 158. Mastocytes lodge particularly around the microvasculature and in the skin and respiratory and gastrointestinal tracts, ready to respond to damages and infections. 159. LPA causes a rapid, reversible, dose-dependent decrease in paracellular flux resistance in brain endothelial cells. LPA modulates tight junction permeability without relocalization of adherens junction-or tight junction-associated proteins. 160. In some types of migrating cells, urokinase-type plasminogen activator receptor (uPAR) associates urokinase-type plasminogen activator (uPA) to the leading edges and controls the activation of small GTPase Rac. As uPAR is not a transmembrane receptor, it interacts with plasmalemmal and cortical proteins such as integrins as well as matrix components such as vitronectin to transmit signals to the cytoskeleton. Melanin-concentrating hormone receptors (Table 7 .77) bind to melanin-concentrating hormone (MCH) that is a nonadecameric cyclic orexinogenic hypothalamic peptide. The latter regulates the feeding behavior, hence energy balance. Melaninconcentrating hormone is generated from a precursor, proMCH, that also produces neuropeptide-EI and -GE. It interacts with neuropeptide-Y [5]. 161 Melanocortin has a protective effect in the cardiovascular system. It attenuates inflammation during myocardial infarction and stroke. The melanocortin receptor fam-161. Melanin-concentrating hormone and α-melanocyte-stimulating hormone (αMSH) have mostly antagonistic, but also agonistic effects. Neuropeptide-EI is a MCH-antagonist and MSH-agonist. Neuropeptide-GE derived from proMCH can mimic MSH signaling via MCHR1 receptor. [1046] . Melanocortin MC 2 receptor on the plasma membrane requires the presence of MRAP1 for its activation. On the other hand, MRAP2 is an endogenous inhibitor, as it competes with MRAP1 for MC 2 binding and decreases the action potency of adrenocorticotropic hormone [1047] . The ACTH hormone indeed binds with high affinity to MC 2 in the presence of MRAP1, but not MRAP2. The influence of MRAP1 and MRAP2 on ligand-binding affinity is specific to MC 2 . The balance between stimulatory and inhibitory accessory proteins controls MC 2 sensitivity to its agonist. These proteins actually have little effect on the binding of α-melanocyte-stimulating hormone to the MC 4 receptor. Thyroid hormones regulate metabolism and appetite. Active triiodothyronine (T 3 ) stimulates the basal metabolic rate. 162 Increased T 3 level prevents transcription of the thyrotropin-releasing hormone (Trh) gene in TRH+ neurons in the paraventricular nucleus of the hypothalamus. These neurons produce both nuclear thyroid hormone receptors 163 and MC 4 melanocortin receptor. Triiodothyronine exerts a negative feedback on MC 4 in hypothalamic paraventricular thyrotropin-releasing hormone neurons [1048] . The MC 4 receptor intervenes in signaling by leptin, a satiety hormone, and links the central energy control to the status of peripheral metabolic reserve. The leptinmelanocortin pathway operates in the hypothalamo-pituitary-thyroid axis to adapt energy expenditure to metabolic reserves. Leptin stimulates TRH production via MC 4 receptor. Activation of MC 4 reduces food intake and increases energy expenditure. Both thyroid hormone receptors ThRα (NR1a1) and ThRβ (NR1a2) con-162. Hyper-and hypothyroidism causes weight loss and gain, respectively. 163. Functional thyroid hormone nuclear receptors (ThRα1, or NR1a1-1, as well as ThRβ1 and ThRβ2, or NR1a2-1 and NR1a2-2) colocalize in many hypothalamic nuclei. Each receptor isoform exerts specific transcriptional activities on the Trh promoter. tribute to MC 4 regulation. The T 3 hormone represses Mc4r gene transcription together with that of Trh gene. It prevents Mc4 expression not only via hypothalamic neurons, but also those of other energy-related brain regions, such as Agouti-related protein-containing neurons [1048] . The T 3 hormone dampens cerebral responsiveness to melanocortin anorectic signaling, but stimulates orexigenic pathways via neuropeptide-Y. Melatonin receptors (MT 1 -MT 3 ) are activated by the endogenous ligands melatonin secreted by the pineal gland to set the circadian rhythm and N acetylserotonin [5] (Table 7 .79). They lodge in the brain and some other organs. Gq/11-coupled motilin receptor (MlnR, MtlR1, or GPR38) is activated by the polypeptide motilin that derives from a precursor that also generates motilin-associated peptide. Motilin is secreted by endocrine M cells in crypts of the small intestine to stimulate gastric activity. Natriuretic peptide receptors include 2 main sets of receptors. Receptors NPR1 and NPR2 are linked to guanylate cyclases (Sect. 6.4.1), whereas NPR3 is a Gprotein-coupled receptor. The NPR3 receptor -the clearance receptor -binds to and removes natriuretic peptides from the blood circulation. It has an extracellular binding domain homologous to that of NPR1 and NPR2, but possesses a truncated intracellular domain that couples via Gi/o proteins to phospholipase-C and inwardly rectifying potassium channels, besides its inhibition of adenylate cyclase. Neuromedin-U receptors (NMU 1 -NMU 2 ; 164 Table 7 .80) are activated by neuromedin-U (NmU). Neuromedin-U is produced at its highest levels in the central ner-164. The NMU 1 receptor was designated as the orphan GPR66 or FM3 and NMU 2 as FM4 or TGR1. vous system, bone marrow, upper gastrointestinal tract, and fetal liver. The structurally related peptide neuromedin-S is also an endogenous agonist of NMU receptors with equivalent potency to that of neuromedin-U [5]. Receptors NMU 1 and NMU 2 couple mainly to Gq/11, but also Gi/o. Receptor NMU 1 is predominantly expressed in peripheral tissues, particularly the gastrointestinal tract, whereas NMU 2 abounds within the central nervous system (brain and spinal cord) [1049] . Neuromedin-U causes vasoconstriction predominantly via NMU 1 and nociception and bone remodeling via NMU 2 . It also prevents obesity. Neuromedin-S is more potent when it binds to NMU 2 receptors in vivo. Like neuromedin-U, it suppresses feeding and contributes to the regulation of the circadian rhythm [1049] . Neuropeptide-B/W receptors (NPBW 1 -NPBW 2 ; Table 7 .81) are activated not only by neuropeptide-B (23 amino acid peptides NPB23) and -W (NPW23), but also their C-terminally extended forms (NPB29-NPW30) [5] . These receptors are predominately produced in the central nervous system. Neuropeptide-S (NPS) is synthesized in the central nervous system. It suppresses anxiety. It targets neuropeptide-S receptor (NPSR; Table 7 .81). Neuropeptide-Y (NPY) receptors (Table 7 .81) are activated by neuropeptide-Y (NPY) and its fragment NPY 3−−36 , peptide-YY (PYY) and its fragment PYY 3−−36 , and pancreatic polypeptide (PP) [5] . Neuropeptide-Y is a sympathetic neurotransmitter involved in the behavior (control of the circadian rhythm and appetite, integration of emotional behavior, and cerebrocortical excitability). Under strong stress, NPY is released with noradrenaline from the peripheral sympathetic nerve terminals to possibly enhance permeability of vascular walls to large molecules, proliferation of vascular smooth muscles, and formation of vascular sprouts in ischemic tissues. Neuropeptide-Y receptors are connected to Gi subunit of heterotrimeric G protein. Five cloned mammalian neuropeptide-Y receptors exist (Y 1 -Y 2 and Y 4 -Y 6 ; Table 7 .81. Receptors for neuropeptide-B (NPB), -W (NPW), -S (NPS) and -Y (NPY), their main G-protein subunit transducers, and ligands (Source: [5] ; GPRA: G protein-coupled receptor for asthma susceptibility; PP: pancreatic polypeptide; PYY: peptide-YY; VRR: vasopressin receptor-related receptor). The Y 3 receptor is the CXCR4 chemokine receptor. Other names Main transducer Potency order Neurotensin receptors (NTS 1 -NTS 2 ; Table 7 .82) are activated by tridecapeptide neurotensin as well as neuromedin-N. Both ligands of neurotensin receptors derive from a common precursor. Receptors NTS 1 and NTS 2 are high-and low-affinity neurotensin receptors, respectively. Receptors of the nicotinic acid family (GPR81 and niacin receptors NiacR1 [or GPR109a] and NiacR2 [or GPR109b]) are targeted by organic acids, such as 165. Receptors Y 1 , Y 2 , Y 4 , and Y 5 are also designated as NPY1R, NPY2R, PPYR1, and NPY5R, respectively. The Y 3 receptor has not been cloned. However, Y 3 receptor is characterized biologically by its inability to be activated by peptide-YY and an ability to be inhibited by NPY 3−36 [1050]. lipid-lowering nicotinic acid (niacin or vitamin-B3). Niacin is converted to nicotinamide and then nicotinamide adenine dinucleotide (NAD) and dinucleotide phosphate (NADP). Therefore, niacin is a precursor to NAD + involved in redox reactions, NADH used as a reducing agent (electron donor), and coenzyme NADP + and its reduced form NADPH. 166 Receptors GPR109a and GPR109b are activated by submicromolar and millimolar concentrations of nicotinic acid, respectively, whereas GPR81 does not respond to nicotinic acid [5] (Table 7 .83). Opioid and opioid-like receptors are activated by various peptides, such as endogenous opioid peptide neurotransmitters Met enkaphalin, Leu enkephalin 167 and βendorphin, 168 as well as active peptides released from prodynorphin cleavage by pro-166. Coenzyme NAD + is reduced to NADH in glycolysis and tricarboxylic acid cycle. NADP + Enzyme cofactor serves as a reducing agent in fatty acid and nucleic acid synthesis. 167. Enkephalins are pentapeptides that contribute to the regulation of nociception. Three different preprohormones contain the enkephalin sequence: preproopiomelanocortin (prePOMC or PPOMC) and preproenkephalin-A and -B (PPEnkA and PPEnkB). 168. β-Endorphin results from the cleavage of proopiomelanocortin that is also the precursor for adrenocorticotrophic hormone. Endorphins (endogenous morphine) are opioid peptides produced by the pituitary gland and the hypothalamus that function as neurotransmitters. The anterior pituitary gland produces the prohormone proopiomelanocortin (POMC). The latter is cleaved into adrenocorticotropin (ACTH) and β-lipotropin. Lipotropin-β is a POMC C-terminal fragment. Lipotropin-β is also involved in lipolysis and steroidogenesis. In particular, hypothalamic histaminergic neurons mediate the ACTH and β-endorphin response to lipopolysaccharides [1052] . protein convertase-2 (α-neodynorphin and dynorphin-A and -B) and prodynorphin partial processing, i.e., big dynorphin, 169 in addition to nociceptin, and tetrapeptides endomorphin-1 and -2 [5] . 170 Endogenous opioids act on plasmalemmal receptors to modulate synaptic transmission. Precursors of endogenous opioid peptides encompass preprodynorphin (PPDyn, PDyn, or PPEnkB), preproenkephalin (PPEnk or PPEnkA; also PEnk or PEnkA), prepronociceptin (PPNoc or PNoc), and preproopiomelanocortin (PPOMC). They are encoded by different genes (PPDYN/PDYN, PPENK/PENK, PPNOC/PNOC, and PPOMC). 171 Prodynorphin (proDyn or PDyn; a.k.a. neoendorphin-dynorphin and preprodynorphin, and proenkephalin-B) is cleaved by proprotein convertase-2 to generate dynorphin-A and -B and α (β)-neoendorphin [1054] . Proenkephalin (proEnk or PEnk), or proenkephalin-A is cleaved into non-opioid synenkephalin (or proenkephalin (1−−70) ), Met enkaphalin, Leu enkaphalin, Met -enkephalin Arg−−Gly−−Leu , Met -enkephalin Arg−−Phe , proenkephalin (114−−133) , proenkephalin (143−−183) , and proenkephalin (237−−258) . The proenkephalin gene is expressed in neurons, hematopoietic (granulocytes, or polymorphonuclear leukocytes, lymphocytes, mastocytes, monocytes, and macrophages) and reproductive cells. Prepronociceptin gives rise to nociceptin (Noc), 172 as well as nocistatin, prepronociceptin (154−−181) , prepronociceptin (169−−176) , Noc2, 173 a nociceptin antagonist in pain transmission, and biologically inactive Noc3 protein. 174 Proopiomelanocortin (POMC) 175 is processed into adrenocorticotropin (ACTH) and β-lipotropin. 176 Lipotropin-β can also be cleaved into smaller peptides, such as γ-lipotropin, β-melanocyte-stimulating hormones (MSH), αto γ-endorphin, and Met enkephalin (Table 7 .84). Four major types of opioid receptors exist: δ-(or Op 1 ), κ-(or Op 2 ), μ-(or Op 3 ), and nociceptin receptors (or Op 4 ; Table 7 .85). δ-Opioid receptors (δ1-δ2 or DOR1-DOR2) are bound by enkephalins. Dynorphin is the primary endogenous ligand 169. Dynorphin inhibits neurotransmitter release at presynaptic terminals. 170. νδoν: within [ νδoγ νης: endogenous, born in the house]; Moρϕ υς: god of sleep and dream. Endomorphins are 2 endogenous opioid peptides of the nucleus of the solitary tract and the periventricular and dorsomedial hypothalamus, especially in hypothalamic histaminergic neurons. 171. Increase of preproopiomelanocortin mRNA in arcuate nucleus and decrease of preprodynorphin mRNA in dentate gyrus of spontaneously hypertensive rats may be associated with the genesis of spontaneous hypertension [1053] . 172. Nociceptin is a heptadecapeptide (content 17 amino acid; πτα: 7; δ κα: 10; πτακαιδ κα: 17) that modulates ion channel activity (hence synaptic transmission) and analgesia. 173. The other heptadecapeptide Noc2 corresponds to the immediately downstream segment from nociceptin in the PNoc precursor. 174. Protein Noc3 corresponds to Noc2 with an added motif of 3 arginine residues (Noc2-Arg-Arg-Arg). 175. Proopiomelanocortin (POMC) is a precursor polypeptide that contains 241 amino acid residues. It is synthesized from a precursor, preproopiomelanocortin (prePOMC), that possesses 285-amino acid residues. 176. Hence its other name corticotropin-lipotropin. . Endogenous opioid peptides include dynorphins, endomorphins, endorphins, and enkephalins. They share the common Nterminal sequence (opioid motif Tyr-Gly-Gly-Phe- [Met or Leu] ). Dynorphins are produced in the hypothalamus, hippocampus, midbrain, medulla, pons, and spinal cord. Endomorphin-1 is widely distributed in the brain and upper brainstem, whereas endomorphin-2 is more prevalent in the spinal cord and lower brainstem. Endorphins (αEnd-γEnd and σEnd) are produced by the hypophysis and hypothalamus. The set of melanocortins includes ACTH, αMSH, βMSH, and γMSH. Proopiomelanocortin, β-lipotropin, corticotropin (ACTH), and corticotropin-like intermediate peptide are secreted by corticotrope cells of the adenohypophysis, or anterior pituitary gland. Opioid peptides . Pre-and postsynaptic μ-opioid receptors (μ1-μ3 or MOR1-MOR3) typically inhibit neurotransmitter release. β-Endorphin has its highest affinity for μ1-, slightly lower affinity for μ2and δ-, and low affinity for κ1-opioid receptors. Endomorphin-1 and -2 have the highest known affinity for μ-opioid receptors. 177 Nociceptin, or orphanin-FQ (OFQ), is an opioid-related peptide that derives from prepronociceptin (PPNoc). Nociceptin binds to nociceptin receptor (a.k.a. κ3-related 177. In the mouse pons and medulla, G-protein activation by endomorphin-1 and -2 is mediated by both MOR1 and MOR2 receptors and that by β-endorphin via MOR2, but not by MOR1 receptors [1058]. G-protein subunit transducers, and ligands (Sources: [5, 1057] ; DOR, KOR, MOR: δ-, κ-, μ-opioid receptor; NOP: nociceptin receptors; TyrMIF: Tyr-Pro-Leu-Gly-NH 2 ; TyrWMIF: Tyr-Pro-Trp-Gly-NH 2 ). Endomorphin-1 and -2 are endogenous opioid peptides highly selective for Op 3 receptor, whereas β-endorphin, another endogenous opioid peptide, targets Op 1 and Op 3 receptors. In general, endogenous opioid receptor ligands modulate effects of various neurotransmitters (acetylcholine, dopamine, noradrenaline, and serotonin) and neurohormones (oxytocin and vasopressin). The majority of opioid peptides undergo a rapid extracellular degradation mainly by integral membrane exoand endopeptidases. Enkephalin is a pentapeptide derived from preproopiomelanocortin and preproenkephalin-and -B ( Met enkaphalin: Tyr-Gly-Gly-Phe-Met [Met in position 5]; Leu Metenkephalin: Tyr-Gly-Gly-Phe-Leu [Leu in position 5]). opioid receptor KORL3, ORL1, OpRL1, OOR, NOCIR, and NOP). This receptor is considered as opioid-related rather than opioid receptor, because, despite its structural homology with conventional opioid receptors, it exhibits a distinct pharmacology. Nociceptin and its receptor NOP lodge in the central and peripheral nervous system, where they modulate nociception [1057] . Nociceptin is, indeed, a potent anti-analgesic. Exogenous nociceptin decreases blood pressure and heart rate via OP 4 receptor in rodents. In the cardiovascular system, OP 4 receptor localizes to sensory afferent fibres, the nucleus tractus solitarius, and rostral ventrolateral medulla, on preganglionic and/or postganglionic sympathetic and parasympathetic nerve fibers innervating blood vessels and heart, as well as in these target organs [1059] . Opioid receptor-δ that belongs to the B-group of rhodopsin subfamily of GPCRs is widely distributed throughout the central nervous system. Opioid Op 1 receptor has also been detected in peripheral tissues (cochlea, cornea, eyelid, and lip, as well as skin fibroblast-like cells, monocytes and T and B lymphocytes [1060] . Endorphins and enkephalins have the highest affinity for Op 1 receptor. The Op 1 receptor can form homodimers as well as heterodimers with other opioid receptors and GPCRs (α2aand β2-adrenergic receptors, CCR5, CXCR2, and CXCR4 chemokine receptors, and sensory neuron-specific receptor SNSR4) [1060] . Agonist-induced monomerization process may be required for receptor endocytosis. It is not only coupled to Orexin-A > orexin-B OX 2 Gq/11 Orexin-A ∼ orexin-B Opioid receptor-κcan regulate the Na + -H + exchange via agonist-induced interactions with Na + -H + exchanger regulatory factor NHERF1 that thus confers cell type-specific signaling to this GPCR. Stimulation of KOR, indeed, activates Na + -H + exchanger NHE3 in cell lines that express high levels of NHERF1, but not other cell types [835] . Orexin receptors are activated by orexin-A and -B 178 that derive from a common precursor, the preproorexin, by proteolytic cleavage [5] (Table 7 .86). Receptors PTH 1 (or PTH1R) and PTH 2 (or PTH2R; Table 7 .87) are activated by parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHRP) as well as related peptides (PTHRP (1−−36) , PTHRP (38−−94) , and osteostatin) [5] . Parathyroid hormone regulates calcium homeostasis and bone metabolism via type-1 PTH receptor. Binding of PTH to PTH 1 (or PTH1R) activates protein kinase-A and -C. Signaling initiated by PTH terminates by PTH 1 endocytosis triggered by β-arrestins. The 178. A.k.a. hypocretin-1 and -2. PTH 1 receptor interacts with Na + -H + exchanger regulatory factor NHERF2 in cells that contain high NHERF levels, such as endothelial cells. In the latter, PTH 1 signals mainly via Gαq and PLCβ, whereas in osteoblasts that do not express detectable levels of NHERFs, PTH 1 transduces signals mainly via Gαs subunit and adenylate cyclase [835] . Scaffold NHERF not only binds to PTH 1 , but also phospholipase-Cβ, protein kinase-C, and transient receptor potential channels that can then reside in close proximity to PTH 1 receptor. The PTH 1 receptor also associates with type-2 transforming growth factor-β receptor TβR2 and forms an endocytic complex in response to PTH [1061] . The TβR2 kinase phosphorylates the PTH 1 cytoplasmic domain to promote PTH-induced endocytosis of the PTH 1 -TβR2-Arrβ2 complex. Platelet-activating factor (PAF, alkyl acetyl-glycerophosphocholine) is a phospholipid mediator that binds to a single G-protein-coupled receptor -PAFR -to activate multiple signaling pathways via Gq/11 and Gi/o family subunits of guanine nucleotide-binding proteins (Table 7 .92). The PAFR receptor may also be activated by lysophosphatidylcholine, oxidized phosphatidylcholine, as well as bacterial lipopolysaccharide [5] . Platelet-activating factor receptor can associate with members of the Janus kinase family of cytosolic protein Tyr kinases such as the TyK2-JaK2 complex composed of tyrosine kinase-2 and Janus kinase-2. Two prokineticin receptors (PK 1 -PK 2 or PKR1-PKR2) are activated by prokineticin-1 (PK1) 179 and -2 (PK2) 180 (Table 7 .88). 181 Prokineticins are widespread. In the cardiovascular system, PK 1 is observed in cardiomyocytes, endothelial cells, and epicardial-derived progenitor cells, 182 but not in vascular smooth muscle cells. Prokineticins potently contract gastrointestinal smooth muscle cells and favor angiogenesis, hence cardiomyocyte survival [1062], among other functions (Table 7.89). In mouse hearts, PK 1 receptor upregulates prokineticins that, in turn, acts 179. Prokineticin-1 was originally named endocrine gland-derived vascular endothelial growth factor (egVEGF). It is also termed mambakine. 180. A.k.a. Bv8. 181. Initially, the peptide hormones prokineticins (PK1-PK2) were identified in the gastrointestinal tract as potent constrictors. They were investigated mostly in the context of angiogenesis in the digestive and reproductive tracts. Subtype PK2 exerts an auto-and paracrine control via PK 1 in the heart and kidney. 182. The actin regulator thymosin-β4 that assists wound repair is able to activate epicardialderived progenitor cells in adults that then differentiate into endothelial cells, smooth muscle cells, and fibroblasts. Prokineticin-2 is also capable of stimulating these progenitors that then differentiate into endothelial and smooth muscle cells, but not fibroblasts. as paracrine factors to promote the differentiation of transcription factor TCF21+ 183 progenitor cells into a vasculogenic cell type [1063] . In isolated renal TCF21+ progenitors, PK2 binds to PK 1 and provokes the differentiation of these progenitors into endothelial and smooth muscle cells. In the reproductive tract, PK1 production rises upon stimulation by estrogen, progesterone, and human chorionic gonadotrophin, as well as hypoxia-inducible factor-1α [1063] . In neurons of the olfactory bulb, PK2 synthesis is primed by basic helixloop-helix (bHLH) transcription factors neurogenin-1 (NeuroG1 or bHLHa6) and achaete-scute complex-like factor ASCL1 (or bHLHa46) 184 and repressed by transcriptional factors distal-less homeobox DLx1 and DLx2. Prokineticins can cause differentiation of human bone marrow cells into the monocyte-macrophage lineage, monocyte proliferation and differentiation, and macrophage migration. In monocytes and neutrophils, PK2 is upregulated in response to granulocyte colony-stimulating factor (CSF3). 183. Transcription factor TCF21 is also called epicardin, capsulin, class-A basic helix-loophelix protein bHLHa23, and podocyte-expressed protein Pod1. 184. A.k.a. mammalian achaete-scute homolog mASH1. The achaete-scute complex is a set of 4 genes (achaete, scute, lethal of scute, and asense) in Drosophila melanogaster that encode basic helix-loop-helix transcription factors. Prokineticin receptors activate many signaling pathways and mobilize calcium messenger. Activated PKRs stimulate PKB, MAPK modules, and the PP3-NFAT axis, as well as phosphoinositol turnover [1063] . In addition, they reduce the production of zonula occludens-1 of tight junctions and can activate protein kinase-C and subsequently transient receptor potential vanilloid TRPV1 channel. Hepatic sinusoidal endothelial cells express only PK 2 receptor. In coronary endothelial cells, PK 1 predominates over PK 2 [1064] . Activated PK 1 that colocalizes with Gα 11 primes mitogen-activated protein kinases and protein kinase-B signaling to stimulate cell proliferation and migration for angiogenesis. Once stimulated by prokineticin-2, PK 1 undergoes endocytosis. On the other hand, prokineticins also cause fenestrations in endothelia [1064] . When PK 2 predominates over PK 1 in coronary endothelial cells, these cells contain a large number of multivesicular bodies and caveolar clusters. Prokineticin-2 activates PK 2 associated with Gα 12 subunit. The latter binds to tight junction protein zonula occludens-1 and trigger its degradation. Prostanoid receptors are activated by various types of prostaglandins, i.e., PGD2, PGE2, PGF2α, PGH2, PGI2 (prostacyclin), and thromboxane-A2 185 (TxA2; Table 7.90). Inflammatory cytokines stimulate the production from arachidonic acid of lipid mediators, such as prostanoids. Rate-limiting enzymes in prostanoid production include cyclooxygenases, 186 in particular constitutively expressed COx1 and inducible COx2. Cyclooxygenase COx2 induced in macrophages and endothelial cells by inflammation stimuli, such as oxidized low-density lipoprotein and interleukin-1, produces eicosanoids that enhance vascular permeability and promote cell chemotaxis. The effects of prostacyclin and prostaglandin-E2 are transduced by the I prostanoid receptor (IP) and E prostanoid receptors (EP), respectively (Table 7 .90; Vol. 5 -Chap. 7. Vessel Wall). Receptors EP 2 and EP 4 are linked to Gs that activates adenylate cyclase, whereas EP 1 and EP 3 are coupled to Gq and/or Gi subunits. Receptors EP 1 and EP 3 mediate PGE2-induced vasoconstriction. Prostaglandin PGE2 can activate platelets by EP 3 . Receptor EP 4 mediates anti-inflammatory effects and activation of MMP9 metallopeptidase. Deletion of EP 2 and IP causes salt-sensitive hypertension. In cardiomyocytes, dominant COx2-derived products are prostacyclin and prostaglandin-E2 that target I prostanoid and E prostanoid EP 2 receptor, respectively. Both PGI2 and PGE2 serves as mediators of cardioprotection. In mice, COx2-deficient cardiomyocytes have impaired ventricular function subsequent to pressure overload [1066] . Although prostanoid receptors are expressed in many of the body's tissues, the distribution of prostanoid receptor types and cell identities that produce each receptor 185. Thromboxanes of the eicosanoid superfamily include thromboxane-A2 and -B2. Thromboxane-A synthase that in platelets converts prostaglandin-H2 to thromboxane. Thromboxane-B2 is an inactive metabolite of thromboxane-A2. 186. There are 3 isoforms of cyclooxygenases, COx1, COx2, and COx3. type is difficult to determine, as prostanoid receptors are expressed at a relatively low level (Table 7 .91). A given cell can synthesize several types of prostanoid receptors. Prostaglandin-D2 is produced by cyclooxygenase-2 that is inducible by mitogens, cytokines, and tumor promoters in activated mast cells, macrophages, and TH2 cells. Prostaglandin-D2 targets receptor DP that is highly expressed by tumor endothelial cells to suppress vascular hyperpermeability via an increase in intracellular cAMP production in tumors [1067] . On the other hand, PGE2 promotes tumor growth and angiogenesis. Prostaglandins PGI2 and TxA2 impede and cause platelet activation and aggregation, respectively. The balance between PGI2 and TxA2 prevents thrombosis and vasospasm, but maintains the body ready to perform efficient hemostasis. Prostaglandins PGI2 and TxA2 are a potent vasodilator and vasoconstrictor, respectively. They abound in vascular endothelial cells and platelets, respectively. However, PGI2 and its main IP receptor are less potent regulators of blood circulation than endothelium-derived vasorelaxant nitric oxide. Prostaglandin-E2 produced by cyclooxygenase-2 can promote relaxation of vascular smooth muscle cells. Prostaglandin PGE2 is synthesized in response to a highsalt diet and works via relaxant EP 2 receptor. Its dysfunction elicits salt-sensitive hypertension. Prostaglandin-E2 EP 1 receptor impairs Na + -Ca ++ exchanges and leads to neurotoxicity [1068] . Membrane-associated proteins involved in eicosanoid and glutathione metabolism (MAPEG) affect the expression of endogenous PGE2 and PGI2. Microsomal prostaglandin-E synthase-1 (mPGES1), a member of the MAPEG family, is a major source of PGE2 formation. Deletion of mPGES1 does not increase blood pressure and retards atherogenesis, whereas mPGES1-derived PGE2 accelerates atherogenesis [1069] . On the other hand, prostacyclin protects the cardiovascular function. In pulmonary arteries, chronic hypoxia increases cyclooxygenase COx2 expression and PGF2α release that activates thromboxane TP receptors [1070] . In pulmonary vascular smooth muscle cells, hypoxia upregulates COx2 expression, thereby increasing production of vasorelaxant (PGI2, PGE2, and PGD2) and vasoconstrictors (PGF2α). The COx1 enzyme, the major TxA2 source, is produced in all wall layers of pulmonary arteries. Its expression remains unaltered in chronic hypoxia. TxA2 synthase that is mainly located in the media of pulmonary arteries is markedly downregulated in chronic hypoxia. In addition, COx enzymes are potential sources of reactive oxygen species in pulmonary arteries, especially in pulmonary arterial hypertension associated with hypoxia. Prostaglandin-F2α activates TP receptors to generate vasocontriction of pulmonary arteries. Whereas cyclooxygenasecatalyzed metabolism of arachidonic acid leads to an endothelium-and COx1dependent relaxation of pulmonary arteries in normoxia, it causes an endotheliumindependent, COx2-and TP receptor-dependent vasocontriction in hypoxia [1070]. In cardiomyocytes, prostaglandins target prostanoid F-receptor (FP) that increases myofilament sensitivity to Ca ++ by enhancing myosin light chain-2 phosphorylation. Prostanoid F-receptor is coupled to Gq/11 protein that leads to production of inositol trisphosphate and diacylglycerol. This second messenger activates PKC, but its effect extends beyond this classical pathway, possiby via G12/13 protein [1071] . Positive inotropic effect of Gq/11-protein-coupled receptors (e.g., α1adrenoceptor, 5HT 2A , and endothelin-1 receptors) results, at least partly, from myofilament Ca ++ sensitization by increased MLC2 phosphorylation. The level of Peptidase-activated receptors (PAR 1 -PAR 4 ; Table 7 .92) are highly expressed in platelets, but also in endothelial cells, myocytes, and neurons. Peptidase-activated receptors are activated by proteolytic cleavage of their N-terminal exodomains by serine peptidases. Among serine peptidases, thrombin and trypsin can be agonist peptidases for certain peptidase-activated receptors in vivo. 187 Alternative endogenous peptidases or ligands to thrombin for PAR 1 , PAR 3 , and PAR 4 exist. Several peptidases, such as cathepsin-G and chymotrypsin, inhibit PAR 1 receptor, as they prevent activation by thrombin [5] . Activated protein-C, 188 an anticoagulant peptidase that elicits an anti-inflammatory response and protects against endothelial barrier dysfunction caused by thrombin, also activates PAR 1 receptor. Therefore, PAR 1 is equipped for peptidase-selective signaling. Tissue factor can also bind peptidase-activated receptor (Table 7 .92). Peptidase-activated receptor PAR 1 is activated by both thrombin and PAR 1 -activating peptide (PAR 1 -AP). Like PAR 1 , PAR 2 on smooth muscle and respiratory epithelial cells can be activated by tryptase, trypsin, and PAR 2 -activating peptide (PAR 2 -AP). 187. Thrombin is produced from prothrombin by factor Xa to mediate the formation of fibrin from fibrinogen that forms the fibrous matrix of blood clots. Thrombin is also a strong activator of platelet aggregation. Moreover, thrombin acts on various cell types, such as monocytes, smooth muscle cells, endothelial cells, fibroblasts, and lymphocytes, among others [1072]. 188. Protein-C on the endothelial cell surface is activated by the thrombin-thrombomodulin complex. Activated protein-C binds to endothelial protein-C receptor (EPCR) and cleaves (inactivates) clotting factors Va and VIIa, thereby reducing thrombin generation. Moreover, it provokes cellular responses via PAR 1 receptor. Four known peptidase-activated receptors are expressed in the entire body (Table 7.93). Thrombin targets mainly PAR 1 , PAR 3 , and PAR 4 , and trypsin chiefly activates PAR 2 (Tables 7.94 and 7.95). Most PARs act via Gi, G12/13 (Ras-Raf pathway), and Gq (PLC-DAG-PKC and PLC-IP 3 -Ca ++ axes) signaling (Table 7 .96). Peptidase-activated receptors participate in the regulation of the vasomotor tone and operate in inflammation, muscle growth, and bone cell differentiation and proliferation. Sorting nexin-1 links to peptidase-activated receptor PAR 1 to favor lysosomal path among endocytosis routes. Thrombin facilitates production and release of growth factors, such as PDGF TGFβ, and ET1, as well as formation of VEGF receptors. Activated PAR 1 participates in synthesis of extracellular factors that contribute to wound healing. Thrombin stimulates procollagen synthesis by smooth muscle cells and pulmonary fibroblasts. It also regulates the release of matrix metallopeptidases, such as MMP1, MMP2, MMP3, and progelatinase-A. PAR 1 Receptor and prothrombin are expressed in numerous regions of the central nervous system, such as thalamus, hypothalamus, cortex, and cerebellum. Thrombin causes increased synthesis of nerve growth factor and endothelin-1. However, in some circumstances, thrombin induces cell death. Thrombin provokes endothelial-dependent relaxation of smooth muscle cells of aortic and coronary arteries via prostacyclin and by nitric oxide. However, in certain territories, it generates vasoconstriction via an increase in intracellular calcium concentration [1072] . Relative expression and function of PAR 1 in endothelial and smooth muscle cells in different vessels can explain the difference in effect on the vasomotor tone. Peptidase-activated receptors PAR 1 and PAR 2 modulate the endothelium-dependent regulation of the vasomotor tone in arteries and veins, such as internal mam- . Although selective PAR 2 -activating peptide (PAR 2 -AP) fails to induce vasorelaxation, PAR 1activating peptide (PAR 1 -AP) generates vasodilation. The endothelium-dependent relaxation is greater in internal mammary artery than in greater saphenous vein. In addition, inflammatory stimuli such as TNFα enhance endothelium-dependent relaxation selectively via PAR 2 -AP in internal mammary artery. Trypsin and peptidase-activated receptor-2-activating peptides, via Ca ++ mobilization, endothelin receptor ET B1 , and subsequent activation of endothelial NO synthase (NOS3), as well as via prostacyclin, are able to cause in vitro an endotheliumdependent (nitric oxide-mediated) relaxation of vascular smooth muscle cells. On the other hand, trypsin and high concentrations of PAR 2 -activating peptides can also initiate endothelium-dependent contraction in both rat pulmonary artery and human umbilical vein [1072] . Whereas endothelium-dependent vasodilation initiated by PAR 2 -APs is due to PAR2 activation, endothelium-independent contraction primed by PAR 2 -APs is done via a receptor different than PAR 2 [1075]. The PAR 1 receptor interacts with Gi/o, Gq/11, and G12/13. The Gi subunit inhibits cAMP and triggers various effects via its activated partner Gβγ dimer. The Gq/11 subunit stimulates phospholipase-C that hydrolyzes phosphoinositides into IP 3 and DAG that increases the cytosolic calcium content and activates protein kinase-C, respectively. Subunits of the G12/13 family activate the RhoGEF-Rho-RoCK pathway for cell remodeling and migration, as well as JNK using small Ras or Rac GTPases. Thrombin also stimulates hydrolysis of other phospholipids via PLA2, PLC, and PLD. The PAR 1 receptor activates Ras via Gi and Tyr kinases after recruitment of adaptors SHC and GRB2, SOS, and Raf1 to the cell cortex then extracellularregulated kinase. The Gβγ subunit activates phosphatidylinositol 3-kinase. Both activated protein-C and thrombin target PAR 1 , but have antagonistic effects, especially on blood coagulation, endothelial barrier permeability, and inflammation. Thrombin excites small GTPase RhoA-associated signaling without Rac1 activation, whereas activated protein-C stimulates small GTPase Rac1, but not RhoA GTPase. Peptidase-selective signaling via PAR 1 relies on caveolin-1, hence PAR 1 compartmentation, as activated protein-C requires caveolin-1, but not thrombin [1076] . Activated protein-C cofactor endothelial protein-C receptor, PAR 1 , Gq and Gi subunits are partitioned into caveolin-1-containing membrane rafts (caveolae). Signal transduction mediated by Gi/o family subunits protects endothelial barrier. In addition, activated protein-C causes PAR 1 phosphorylation. It also desensitizes endothelial cells to thrombin. Moreover, it limits receptor proteolysis, its endocytosis, and degradation, even after prolonged exposure to activated protein-C. The PAR 2 receptor on both endothelial cells and leukocytes intervenes in inflammation. Trypsin and PAR2-activating peptides stimulate activation of T lymphocytes and neutrophils, and promote leukocyte recruitment (rolling, adhesion, and extravasation) by a mechanism dependent on platelet-activating factor release [1077]. The PAR 3 receptor resides in heart, small intestine, and bone marrow, among other organs, especially on airway smooth muscle cells, vascular endothelial cells, and astrocytes. In mouse platelets, PAR 3 expression is necessary for full activation by thrombin. Tissue distribution of PAR 4 differs from that of other PARs, with the highest levels in the lung, small intestine, pancreas, thyroid, and testis. The PAR 4 receptor operates as a low-affinity thrombin receptor. Tissue factor can have 2 distinct structural and functional modes, as it is an initiator of blood clotting, 189 or a cofactor for cell signaling that is unable to promote coagulation. 190 In other words, plasmalemmal tissue factor binds to: (1) serine peptidase factor VIIa to activate coagulation or (2) peptidase-activated receptor-2 to trigger inflammation and angiogenesis, as tissue factor belongs to the cytokine receptor family. 191 Tissue factor-PAR 2 complex formation can be inhibited without preventing the coagulation activity of tissue factor. Protein disulfide isomerase (PDI) 192 stabilizes a distinct tissue factor-factor VIIa complex that does not bind factor X [1078]. PDI inhibits the coagulation activity of the tissue factor, and switches tissue factor to cell signaling. 193 Inflammation and coagulation initiated by tissue factor are coupled by a crosstalk between peptidase-activated receptor-1 and sphingosine 1-phosphate receptor-3, at least in dendritic cells [1079] . Signaling from PAR 1 is also coupled to S1P in endothelial cells. Dendritic cells in the lymphatics can cause exacerbated inflammation and coagulation in deregulated innate immune responses such as disseminated intravascular coagulation. Block of PAR 1 is then sufficient to interrupt systemic dissemination of inflammation and coagulation. 189. Tissue factor, an integral membrane protein, is normally excluded from the endothelial wetted surface. Tissue factor also circulates and becomes active only with a growing thrombus. Coagulation (Vol. 5 -Chap. 9. Endothelium) is triggered when tissue factor is exposed to zymogen coagulation peptidases in plasma after vessel wall damage. It binds and activates factor VII. The complex made of coagulant tissue factor and factor VIIa binds and activates factor X. Factor Xa acts with its cellular cofactor Va for thrombin production. Thrombin cleaves fibrinogen to generate fibrin and causes platelet aggregation. 190. Tissue factor is also activated in inflammation, vascular development, and cancer. 191. The non-coagulant form of tissue factor bound to factor VIIa (binary signaling complex) activates receptor PAR 2 . Factor Xa signals via peptidase-activated receptors PAR 1 and PAR 2 either as a monomer or as a ternary complex with tissue factor and factor VIIa. Factor Xa is inhibited by antithrombin-3 and tissue factor pathway inhibitor. Factor VIIa is also inhibited by tissue factor pathway inhibitor bound to factor Xa. Thrombin is a potent activator of peptidase-activated receptors, except PAR 2 receptor. It activates PAR 1 and PAR 4 for platelet aggregation. Like Factor Xa, thrombin is inhibited by antithrombin-3. Thrombomodulin hampers the binding of thrombin with PAR 1 on the endothelial cell surface. 192. Protein disulfide isomerase cleaves disulfide bonds in the extracellular domains of certain receptors to regulate protein activity. Protein disulfide isomerase breaks a disulfide bond that is required to activate the coagulation. Nitric oxide regulates PDI activity, hence, suppresses the coagulant activity of tissue factor. 193. Disulfide/thiol exchange is required for the formation of Tissue factor-PAR 2 complex. LGR: leucine-rich repeatcontaining G-protein-coupled receptor; Rln3R: relaxin-3 receptor; SALPR: somatostatin and angiotensin-like peptide receptor). The Gα oB subunit is a member of the Gα i/o family (with Gα oA , Gα i1 , α i2 , α i3 , Gα Tr , and Gα z ). Other names The set of relaxin family peptide receptors are divided into 2 categories: group 1 (RXFP 1 -RXFP 2 ) and 2 (RXFP 3 -RXFP 4 ; Table 7 .97). They are targeted by heterodimeric peptide hormones relaxin-1 to -3 and insulin-like peptide InsL3 and InsL5 [5] . Relaxin receptors reside in central and autonomous nervous system, heart, smooth muscle, and connective tissue. At least 2 binding sites, with high and low affinity, exist on RXFP 1 and RXFP 2 receptors [5] . The RXFP 1 receptor activates adenylate cyclase, protein kinase-A and -C, phosphatidylinositol 3-kinase, and extracellular signaling regulated kinases ERK1 and ERK2 [1080] . It also intervenes in nitric oxide signaling. Relaxin targets RXFP 1 for connective tissue remodeling. Gs-Coupled RXFP 2 activates and Gi-coupled RXFP 3 and RXFP 4 and inhibit adenylate cyclase, respectively. The RXFP 3 receptor also activates ERK1 and ERK2 kinases. Except ionotropic receptors of the 5HT 3 class that are ligand-gated Na + and K + cation channels, all other serotonin (or 5-hydroxytryptamine) receptors (5HT 1 -5HT 2 and 5HT 4 -5HT 7 ; Table 7 .98) are G-protein-coupled receptors. The diversity of metabotropic 5HT receptors is augmented by alternatively spliced variants. Subtypes 5HT 2A , 5HT 2C , and 5HT 6 are non-functional [5] . Furthermore, RNA editing produces 5HT 2C isoform that differ in efficiency and specificity of Gq/11 coupling. Serotonin metabotropic receptors reside in the central and peripheral nervous system, where they activate an intracellular cascade to produce an excitatory or inhibitory response (Table 7 .99), as well as other tissues, particularly vasculature. The bioamine serotonin is synthesized from tryptophan. Once released, serotonin increases the smooth muscle tone. The central serotoninergic system innervates diverse brain regions. In the central nervous system, cell bodies that contain serotonin localize to nuclei raphé, near the midline of the brainstem, and project to the brainstem and spinal cord as well as forebrain [1081] . The activity of the raphé network is modulated by activators and inhibitors of the release of serotonin. Various serotonin receptors modulate the synaptic transmission and postsynaptic excitability. Serotonin receptors indeed influence the release of many neurotransmitters, such as acetylcholine, adrenaline, dopamine, GABA, glutamate, and noradrenaline, as well as many hormones, such as corticosterone, corticotropin, cortisol, oxytocin, prolactin, substance-P, vasopressin, etc. Disc large homolog DLg4 194 connects to 5HT 2A and 5HT 2C to facilitate their anchoring and clustering to postsynaptic dendrites and agonist-dependent internalization, respectively [835] . Breathing is controlled by the nervous system. The serotoninergic apparatus belongs to the arousal system that controls the body's ventilation. Adjustment of excitability of the ventilatory command is also regulated by acetylcholine, adenosine, catecholamines, opioids, and other neuropeptides [1081] . Apneusis consists of a pause at inspiration and prolonged breath holding. Central apnea results from the loss of inspiratory motions. 195 Serotonin receptors can counteract both apneusis and opioid-induced apnea [1081] . In mammals, the respiratory rhythm is generated by the pre-Bötzinger complex (PBC), a cluster of interneurons in the ventrolateral medulla of the brainstem. Many neurons of the pre-Bötzinger complex produce adenosine (A 1 and A 2A ), GABA, glutamate, neurokinin-1 (NK 1 ), and serotonin receptors, such as 5HT 1A , 5HT 2A , 5HT 2B , 5HT 4 , and 5HT 7 . Serotonin receptors colocalize with opioid Op 3 receptor in respiratory neurons of the pre-Bötzinger complex to promote activity of neurotrophins. Gq-Coupled 5HT 2A and 5HT 2B receptor isoforms are coexpressed in these neurons. They does not excite breathing. Only 5HT 1A and 5HT 4 isotypes are able to counteract opioid-induced respiratory depression [1081]. 194. A.k.a. PSD95. 195 . These respiratory disorders can be caused by brainstem tumors or degeneration such as olivopontocerebellar hypoplasia, as well as opioid or barbiturate administration. Table 7 .98. Subtypes of serotonin receptors and their direct functions in the circulatory and ventilatory systems as well as via the central (CNS) and peripheral (PNS) nervous system (Source: Wikipedia). Among the 7 categories of 5HT receptors (5HT 1 -5HT 7 ), 1 ionotropic serotonin receptor (5HT 3 transmitter-gated Na + -K + ion channel), and 15 different metabotropic isoforms have been identified. In the central nervous system, Gi-coupled 5HT 1A is predominantly expressed in the hippocampus, lateral septum, and brain cortex, as well as in brainstem regions, such as nuclei raphé and hypoglossal nuclei. On presynaptic terminals, it precludes presynaptic transmitter release. At postsynaptic membranes, it activates via Gβγ subunit inwardly rectifying K + channels and inhibits Ca ++ channels. In the central respiratory network, serotonin lowers excitability of respiratory neurons [1081] . Activated 5HT 1A receptor enhances opioid-induced bradycardia, but partially compensates the decrease in vascular resistance and sympathetic activity, in addition to breathing restoration [1082]. The Gi/o-coupled 5HT 1B receptor controls several signaling axes [1083]: (1) inhibition of adenylate cyclase; (2) inhibition of Rap1 guanine nucleotide-exchange factor (Rap1GAP); (3) activation of phospholipase-C and stimulation of Ca ++ release; (4) phosphorylation of extracellular signal regulated kinase, phosphatidylinositol 3-kinase, P70 ribosomal S6 kinase (S6K), and protein kinase-B; (5) activation of Ca ++ -dependent K + channels; and (6) stimulation of nitric oxide production. It is primarily expressed in the central nervous system, both pre-and postsynaptically (on serotoninergic axon terminals), where it regulates the release of various neurotransmitters (glutamate, dopamine, GABA, acetylcholine, and N acetyl aspartyl glutamate), including serotonin itself (e.g., terminal autoreceptors of serotoninergic neurons) [1083] . It also lodges in sympathetic nerves of the gastrointestinal tract. The 5HT 1B receptor is also synthesized in smooth muscle cells of cerebral arteries and other blood vessels. It impedes noradrenaline release in the vena cava as well as trigeminal ganglion-stimulated plasma extravasation in rodents [1083] . It mediates the vasoconstriction of rat caudal arteries. The 5HT 1B receptor can homo-and heterodimerize with 5HT 1A and 5HT 1D . It can interact with GSK3β, glutathione S-transferase-κ1, and non-metastatic cell subunit NME2 of nucleoside diphosphate kinase. It is internalized and interacts with various regulators. Most activated GPCRs associate with arrestin. A single GPCR can then trigger multiple signaling events according to the presence or absence of β-arrestin and the type of binding agonist. Different patterns of signal transduction and functional selectivity of 5HT 2A receptor 196 is determined by β-arrestin. The absence of β-arrestin-2 abrogrates many effects of serotonin from 5HT 2A receptor, but has no or little effect on the same signaling pathways with other kinds of agonists. In the central nervous system, 5HT 2B receptor 197 participates in embryo-and fetogenesis. It is also involved in anxiety, migraine, schizophrenia, autism, and depression [1081]. The 5HT 2C receptor (previously called 5HT 1C ) is coupled to Gα q , hence to phospholipase-Cβ. It is exclusively expressed in the central nervous system. It is the single GPCR that undergoes adenosine-to-inosine RNA editing at 5 positions, hence generating multiple functional variants with different G-protein-coupling properties and transfer modes [1084] . 198 The 5HT 2C receptor intervenes in various behavioral and physiological processes, such as regulation of mood, nociception, motor behavior, endocrine secretion, thermoregulation, modulation of appetite, cerebrospinal fluid production by choroid plexi, and control of exchanges between the central nervous system and cerebrospinal fluid [1084] . It contributes to the control of mesocorticolimbic and nigrostriatal dopaminergic systems, as it inhibits dopamine release. The 5HT 2C receptor has both constitutive and inducible activity [1084] . Desensitization of its agonist-dependent activation results from receptor phosphorylation by G-protein-coupled receptor kinase GRK2 that is followed by recruitment of βarrestins, which uncouples 5HT 2C from G protein and promotes its internalization into endosomes. Endocytosis not only ensures receptor desensitization, but also allows receptor dephosphorylation and recycling to the plasma membrane for resensitization. Owing to transactivation of the small GTPase RhoA, 5HT 2C receptor can activate phospholipase-D. It can also stimulate phospholipase-A2 and extracellular signalregulated kinases ERK1 and ERK2. It also interacts with phosphatase and tensin homolog. 196. In the central nervous system, 5HT 2A , preferentially coupled to Gq localizes to the neocortex, entorhinal and pyriform cortex, claustrum, caudate nucleus, nucleus accumbens, olfactory tubercle, hippocampus, and cerebellum [1081] . 197. The 5HT 2B receptor resides in the neocortex, cerebellum, dorsal hypothalamus, and medial amygdala [1081] . 198. Pre-mRNA editing generates multiple functional variants with decayed constitutive activity, agonist affinity, and PLC activation potency, as well as additional G13-coupling capacity, but modified mediator selectivity and trafficking. Sphingosine 1-phosphate is a lipid growth factor (Vol. 2 -Chap. 3. Growth Factors) that acts via its specific G-protein-coupled receptors (Table 7 .101). Sphingosylphosphorylcholine (SPC) also activates this receptor with equal, smaller, or greater potency than that of sphingosine 1-phosphate according to the receptor type. The S1P receptor family comprises 5 receptors (S1P 1 -S1P 5 ) that regulate cell survival, proliferation, and migration. Sphingosine 1-phosphate receptors are members of the endothelial differentiation gene (EDG) family with lysophosphatidic acid receptors. Receptors S1P 1 to S1P 3 are widespread, whereas S1P 4 and S1P 5 reside in cells of the nervous and immune systems. Endocytosis of S1P GPCRs can trigger Gi-mediated signaling similarly to that initiated from the plasma membrane [11]. The major S1P 1 receptor is observed on vascular endothelial and smooth muscle cells. It couples to Gi protein. It regulates endothelium-dependent vasorelaxation, smooth muscle contraction, and blood vessel maturation during embryo-and fetogenesis [1085] . It also controls the transfer of immunocytes. Lipidic S1P ligand is predominantly stored and released by erythrocytes. It is present at high concentrations (100 nmol-1 μmol) in the blood and lymph. A large S1P fraction is bound to plasma lipoproteins and albumin. Upon S1P binding, S1P 1 receptor primes egress of lymphocytes from secondary lymphoid organs toward the blood (low S1P concentration) or lymph (high S1P concentration) [1086] . In endothelial cells, transient exposure of exogenous S1P 1 ligand 204 triggers ligand-bound S1P 1 endocytosis and long-lasting signaling with inhibition of adenylate cyclase and increased ERK phosphorylation that causes augmented cell migration, whereas calcium response is abrogated [1086] . On the other hand, S1P agonist does not provoke persistent signaling from internalized S1P 1 receptors. Widespread S1P 2 couples to Gi, Gq, and G12/13 types of Gα subunit of heterotrimeric G-proteins to regulate several mediators, such as Rho GTPase, PTen phosphatase, ERK kinase, and VE-cadherin (Table 7 .102). However, it preferentially activates G12/13 family subunits. Activated S1P 2 on endothelial cells provokes disruption of VE-cadherin-based adherens junctions, hence increasing endothelial paracellular permeability [1085] . Because S1P 2 activates RhoA, it impedes Rac activity. The S1P 2 receptor actually inhibits small GTPase Rac, as it stimulates a GTPase-activating protein for Rac GTPase, thereby impeding cell migration. In addition, S1P 2 stimulates cAMP synthesis via G13 (but not Gs). Once coupled to G12/13 family subunits, it can also activate monomeric Rho GTPase. The S1P 2 receptor is able to activate the Gi-Ras-MAPK axis to regulate cell proliferation and survival. Phosphorylation of ERK kinases, in turn, stimulates Jun and Fos transcription factors. In addition, S1P 2 activates other members of the MAPK 204. E.g., immunomodulator drug fingolimod. ). Five known receptors bind to S1P ligand. Receptors S1P 1 to S1P 3 are widespread; S1P 4 and S1P 5 reside in blood cells. In cells of the cardiovascular apparatus, S1P 1 couples exclusively to Gi protein; S1P 2 and S1P 3 to subunits of the Gi, Gq, and G12/13 families; and S1P 4 and S1P 5 to those of the Gi and G12/13 families. The Gi subunit inhibits adenylate cyclase, whereas Gβγ can activate ion channels and kinases; Gq subunit targets phospholipase-C; and G12/13 subunit connects to RhoA guanine nucleotide-exchange factor. Because Gi protein prevents cAMP formation, it precludes PKA-mediated activation of Ca V 1 channel. The Gβγ dimer associated with Gi acts on Gi-regulated inward rectifier K IR 3 channel (K ACh current) that contributes to resting membrane potential in human atriomyocytes. Cardiomyocytes S1P 1 S1P 3 > S1P 2 Cardiac fibroblasts S1P 3 S1P 1 > S1P 2 Aortic smooth muscle cells S1P 2 > S1P 3 S1P 1 Vascular endothelial cells S1P 1 > S1P 3 S1P 2 family -JNK and P38MAPK -that operate in response to cell stress, in particular for cytokine production. It also activates the Gq-PLC-Ca ++ pathway as well as phospholipase-D. Vascular endothelial and smooth muscle cells as well as cardiomyocytes and cardiac fibroblasts express S1P 1 to S1P 3 receptors, but with different expression patterns (Table 7 .103). Because S1P 1 is the most abundant type in endothelial cells and cardiomyocytes, most S1P-primed responses occur via Gi-coupled S1P 1 receptor alone or in combination with Gi-, Gq-, or G12/13-coupled S1P 3 receptor. In vascular smooth muscle cells, S1P 2 has the highest density. Stimulated S1P 1 or S1P 3 activates Rac GTPase, whereas stimulated S1P 2 inhibits Rac GTPase. Cardiac fibroblasts express predominantly S1P 3 receptor. In addition, sphingosine kinase activity is higher in cardiac fibroblasts than in cardiomyocytes [1087] . In cardiomyocytes, S1P 1 receptor inhibits cAMP formation and antagonizes adrenergic receptor-mediated inotropy [1087] . The S1P 1 receptor localizes to caveolae in ventriculomyocytes. The S1P 3 receptor induces bradycardia. Both S1P 2 and S1P 3 receptors favor cardioprotection in ischemia-reperfusion injury. Receptors of S1P also participate in remodeling, proliferation, and differentiation of cardiac fibroblasts. In endothelial cells, caveolar domains of the plasma membrane are enriched in NOS3 and contain S1P 1 receptors. Lipid S1P activates NOS3 via: (1) Ca ++calmodulin that relieves caveolin inhibition and (2) protein kinase-B that phosphorylates NOS3. Sphingosine 1-phosphate enhances the endothelial barrier. In particular, it prevents the permeability increase caused by platelet-activating factor or bradykinin. Among S1P 1 to S1P 3 located on endothelial cells, only S1P 1 protects the endothelial barrier against the effect of platelet-activating factor and bradykinin in rat venules [1088] . Sphingosine 1-phosphate stabilizes newly formed vessels and antagonizes thrombin that disrupts the endothelial barrier via S1P 1 and S1P 3 receptors [1087] . It interacts with S1P 1 during angiogenesis and vasculature maturation [1089] . Gi-Coupled S1P 1 receptor regulates cell survival, proliferation, migration, and morphogenesis in response to S1P messenger. Activated S1P 1 also stimulates the assembly of cadherin complexes in endothelial cells. In endothelial cells, S1P 1 activates α V β 3 -and β 1integrins via GTPase Rho [1090] . Liganded S1P 1 primes the Gi-PI3K-PKB, PI3K-Rac, PLC-DAG-PKC and -IP 3 -Ca ++ , and Ras-ERK pathways. Protein kinase-B then binds to and phosphorylates S1P 1 , hence regulating GTPase Rac and cortical actin assembly, lamellopodium formation, and chemotaxis [1091] . Activated S1P 1 favors cell survival and proliferation of vascular endothelial cells and stimulates nitric oxide synthase in endothelial cells via Gi and PI3K [1092] . Whereas S1P 2 inhibits Rac and growth factor-induced chemotaxis [1093] , S1P 3 activates small GTPase Rho. In vascular smooth muscle cells, S1P activates Rho GTPase and promotes myosin light-chain phosphorylation that provokes vasoconstriction mainly via S1P 3 receptor. On the other hand, S1P can promote the formation of nitric oxide in endothelial cells that, once released, relaxes smooth muscle cells. Sphingosine 1-phosphate and lysophosphatidic acid are produced by mastocytes, platelets, and macrophages. Ligands S1P and LPA affect immunocyte survival, differentiation, proliferation, migration, receptor expression, and protein synthesis and secretion (Table 7 .104). The emigration of thymocytes from the thymus, transfer of lymphocytes between blood and secondary lymphoid organs (thus avoiding sequestration in lymph nodes as well as persistence in blood), and immigration of B lymphocytes into splenic follicles (white pulp of spleen) is assisted by S1P and its receptors [1094] . Sphingosine 1phosphate in dendritic and T cells enhances the IgE production. In an autocrine manner, via interactions with different GPCRs, S1P enhances mastocyte migration and release of pro-inflammatory mediators in allergy. Whereas LPA promotes mastocyte development, S1P-bound S1P 1 increases mastocyte chemotaxis. In contrast, chemotaxis of mastocytes is prevented by IgE-upregulated S1Pbound S1P 2 receptor. In addition, S1P 2 augments IgE-mediated release of allergic mediators. Tachykinin receptors are activated by members of the tachykinin family of neuropeptide neurotransmitters: substance-P, neurokinin-A 205 and -B, 206 and neuropeptide-K, 207 and -γ. 208 Three distinct tachykinin receptors have been identified (NK 1 -NK 3 , or NK1R-NK3R). Substance-P preferentially activates NK 1 , neurokinin-A NK 2 , and neurokinin-B NK 3 (Table 7 .105). In the respiratory tract, both NK 1 and NK 2 receptors are 205. A.k.a. substance-K, neurokinin-α, and neuromedin-L. 206. A.k.a. neurokinin-β and neuromedin-K. 207. A.k.a. neurokinin-K. 208. The human tachykinin genes TAC1, TAC3 (mouse TAC2 gene), and TAC4 produce tachykinins, as they encode tachykinin precursors, the preprotachykinins that undergo proteolytic cleavage to form smaller peptides, and multiple splice variants that leads to different sets of peptides. Each preprotachykinin type is the product of one gene. The TAC1 gene encodes preprotachykinin-A (PPTKa; a.k.a. protachykinin-1 and tachykinin precursor-1), leads to formation by alternative splicing to neurokinins-A and -K and neuropeptide-γ. All TAC1 splice variants (α, β, and γ splice variants) manufacture substance-P, but only β and γ splice variants produce neurokinin-A, neuropeptide-K, and neuropeptide-γ. Neuropeptide-K and -γ are versions of neurokinin-A with a longer N-terminus. The genes TAC3 and TAC4 encode preprotachykinins-B (PPTKb) and -C (PPTKc) that produce neurokinin-B and hemokinin-1, respectively. Trace amine receptors, or trace amine-associated receptors, (TA 1 -TA 2 ) are bound by trace amines (Table 7 .106). Trace amines are endogenous compounds that are structurally related to classical biogenic amines. They are produced in the nervous system. They colocalize in neurons in which these biogenic amines serves as neurohormones, neuromodulators, and/or neurotransmitters, such as monoamines that encompass catecholamines, which derive from phenylalanine and tyrosine 210 (adrenaline, dopamine, and noradrenaline), serotonin, histamine, and melatonin. Trace amines include: (1) tyramine, 211 a monoamine derived from tyrosine; 212 (2) phenylethylamine (PEA), a natural monoamine alkaloid that is synthesized from 209. Tachykinin receptors can also be found in nerves (NK 1 ) and inflammatory cells (NK 2 ), such as T lymphocytes, macrophages, and mastocytes [1095] . 210. Tyrosine is either generated from phenylalanine by phenylalanine hydroxylase or processed from ingested proteins. 211. A.k.a. 4-hydroxyphenethylamine and para-tyramine. 212. Tyramine releases catecholamines (adrenaline, dopamine, and noradrenaline). It is metabolized by monoamine oxidase. A large dietary intake of tyramine can cause the tyramine pressor response, with an increase in systolic blood pressure associated with vasoconstriction, but repeated exposure to tyramine reduces tyramine pressor response. (3) tryptamine, another monoamine alkaloid; 213 (4) octopamine; 214 (5) 3-iodothyronamine that has negative inotropic and chronotropic effects [1097]; and (6) psychostimulant amphetamines 215 (α-methylphenethylamines). Endogenous amphetamines, like all trace amines, bind to trace amine receptors, or trace amine-associated receptors. The TA 1 receptor localizes to the central nervous system as well as peripheral organs such as the kidney. It may be targeted by thyronamines, endogenous derivatives of thyroid hormones (decarboxylated and deiodinated metabolites). Thyrotropin-releasing hormone receptors (Table 7 .107) are activated by the endogenous tripeptide thyrotropin-releasing hormone (TRH) that is produced by medial neurons of the hypothalamic paraventricular nucleus to stimulate the release of thyroid-stimulating hormone and prolactin by the adenohypophysis (endocrine anterior lobe of the pituitary gland). Urotensin-2 receptor 216 is a Gq/11-coupled receptor. In humans, its highest levels are detected in skeletal muscle and cerebral cortex and lower levels in kidney cortex and left ventricle [1098] . Urotensin-2 is synthesized from preprourotensin-2 isoforms that contain 124 and 139 amino acids. Urotensin-2 causes vasoconstriction in human arteries (e.g., coronary, mammary, and radial arteries) and veins (e.g., 213 . Tryptamine serves as a backbone for members of the tryptamine group. Tryptamine derivatives comprise neurotransmitter serotonin (5-hydroxytryptamine) and sleep-wake cycle regulator neurohormone melatonin ( N acetyl 5-methoxytryptamine). Serotonin is synthesized from tryptophan (α-carboxyltryptamine) successively by tryptophan hydroxylase and amino acid decarboxylase. Serotonin, in turn, can be converted to melatonin by N acetyltransferase and 5-hydroxyindole O methyltransferase. 214. Tyramine is hydroxylized to octopamine by dopamine β-hydroxylase to be subsequently packaged in synaptic vesicles with noradrenaline. In mammals, octopamine may release lipids from adipocytes. Whereas serotonin increases peristaltic movements in the digestive tract, octopamine suppresses them [1096] . 215. Endogenous amphetamines are manufactured in the central and peripheral nervous system. They modulate the level of excitement and alertness. 216. A.k.a. hypocretin receptor; UTR, UTR2, UTs2R, or GPR14. Magnocellular neurosecretory cells of the paraventricular and supraoptic nuclei of the hypothalamus synthesize one of the neurohypophysial hormones -nonapeptides arginine vasopressin (AVP), 217 or antidiuretic hormone (ADH), and oxytocin (OT) -along with different neuropeptides or neuromodulators. Vasopressin and oxytocin receptors are activated by vasopressin and oxytocin. Vasopressin signals via 3 cognate receptors (Tables 7.108 and 7.109). Oxytocin receptor requires Mg ++ ion and cholesterol. Hormones oxytocin and vasopressin not only act on peripheral organs via the blood circulation, but are also released in the brain, where they influence social be-217. Lysine vasopressin (LVP), or lypressin, plays the same role in pigs. havior. 218 Vasopressin contributes to water reabsorption in the renal collecting ducts via aquaporin-2. Vasopressin also causes a moderate vasoconstriction. Neuropeptide oxytocin acts as a hormone with several peripheral actions and a neurotransmitter in the central nervous system released from centrally projecting oxytocin neurons. Oxytocin intervenes in parturition initiation and milk ejection during lactation, in addition to psychosocial behavior. Oxytocin and vasopressin are stored in vesicles in and released by the neurohypophysis into the blood circulation. 219 Oxytocin is synthesized as an inactive precursor that includes its carrier neurophysin-1. 220 It is stored with neurophysin-1 in Herring bodies at axon terminals in the neurohypophysis. Vasopressin and its carrier neurophysin-2 also derives from the same precursor. 221 Regulation of water transport across the epithelium of the collecting duct of the nephron enables a precise control of renal water excretion, thereby regulating the osmolality of body fluids. Water transport control is mainly achieved by vasopressin that binds to V 2 receptor in the basolateral plasma membrane of principal cells of collecting duct to cause an increase in intracellular cAMP and Ca ++ concentrations. Vasopressin, indeed, favors via V 2 the transfer of aquaporin-2 to the apical plasma membrane, as it triggers phosphorylation by myosin light-chain kinase of the myosin regulatory light chain, as well as aquaporin-2 phosphorylation (Ser256, and Ser264, and S269) and dephosphorylation (Ser261) [1101] . Vasopressin primes activation of several kinases of the AGC family, such as cAMP-dependent protein kinase-A and calmodulin-dependent kinase-2 (via autophosphorylation at Thr286), but lowers phosphorylation of cyclin-dependent kinases and members of the mitogen-activated protein kinase family, such as JNK1 and JNK2 (Thr183 and Tyr185) and ERK1 and ERK2 (Thr183 and Tyr185; Thr203 and Tyr205). In normal circumstances, vasopressin attenuates activities of ERK1 and -2 and JNK1 and -2, but not P38MAPK. Aquaporin-2 is strongly phosphorylated (Ser261) by JNK, P38MAPK, and CDK5 and -9, as well as weakly by ERK kinases. It is phosphorylated (Ser256) by protein kinase-A. 218 . In rats, vasopressin signals in the olfactory system for proper social recognition [1102] . A population of vasopressin neurons resides in the olfactory bulb. In humans, oxytocin participates in the processing of social stimuli [1100] . Oxytocin reduces activation in lateral and dorsal regions of the anterior amygdala in the presence of negative social stimuli, but enhances activity for positive social stimuli. Furthermore, oxytocin increases activity in the posterior amygdala and promotes the coupling of this subregion to the superior colliculi [1100] . Oxytocin also operates in behavioral and endocrine stress responses. It contributes to increase trust and reduce betrayal aversion. 219. The pituitary gland consists of adeno-(anterior pituitary) and neurohypophysis (posterior pituitary). The pituitary secretes hormones that regulate water homeostasis, blood pressure, development, and reproduction. 220. Neurophysin-1 and oxytocin result from the cleavage of a common precursor: preprooxyphysin, or preprooxytocin-neurophysin-1. 221. Neurophysins also act as neurohypophysial hormone. Neurophysin-2 stimulates prolactin secretion. It is generated from the same precursor as vasopressin: pre-proArg vasopressin-neurophysin-2. GRF: growth hormone-releasing factor; PHI: peptide histidine isoleucineamide). Two -short and long -PACAP isoforms (PACAP27 and PACAP38) derive from proteolytic cleavage of a precursor (proPACAP(131-157) and proPACAP(131-168), respectively). Vasoactive intestinal peptide and pituitary adenylate cyclase-activating peptide receptors are activated by endogenous peptides vasoactive intestinal peptide (VIP), pituitary adenylate cyclase-activating peptides PACAP27 and PACAP38, peptide histidine isoleucineamide (PHI), peptide histidine methionine amide (PHM), peptide histidine valine (PHV), and growth hormone-releasing factor (GRF) [5] . Vasoactive intestinal peptide causes smooth muscle relaxation, exo-and endocrine secretion, and water and ion flux in respiratory and intestinal epithelia. Pituitary adenylate cyclase-activating polypeptides are neuropeptides that belong to the VIP-PACAPglucagon-secretin family. They can operate as neurotransmitters and neuromodulators. In addition, they contribute to para-and autocrine regulation of some cell types. The VIP-PACAP receptors constitute a family (VPAC 1 , VPAC 2 , and PAC 1 ; Table 7.110). The PAC 1 receptor is expressed in brain, adrenal medulla, pancreatic acini, uterus, and myenteric plexus [1106] . The VPAC 1 receptor is widely distributed, but predominantly detected in the lung, small intestine, thymus, and, within brain, in the cerebral cortex and hippocampus [1103] . (It is also found in the heart, kidney, liver, spleen, colon, prostate, testis, and placenta, as well as T lymphocytes [1104] ). The VPAC 1 receptor is also activated by secretin. The VPAC 2 receptor is also broadly distributed (central nervous system, skeletal and smooth muscles, heart, kidney, adipose tissue, pancreas, 222 stomach, and testis). In the central nervous system, VPAC 2 receptor is present in regions associated with neuroendocrine function, such as several hypothalamic nuclei. It is also detected in the pituitary gland and pancreatic islets [1103] . In the human respiratory tract, VPAC 2 is observed in tracheal and bronchial ciliated epithelial cells as well 222 . It has been isolated from a mouse insulin-secreting β-cell line. as mucous and serous cells of submucosal glands, bronchiolar epithelial cells, and alveolar macrophages, but not airway and vascular smooth muscle and endothelial cells [1105] . Gs-coupled 5HT 4 receptor 199 contributes to the control of transmitter secretion and cognition. It localizes also to the myocardium, adrenal glands, digestive tract (from esophagus to colon), and bladder Types 5HT 4A and 5HT 4B couple also to G13 and Gi/o family subunit, respectively. Unlike 5HT 1A , 5HT 4A receptor further reduces the decrease in opioid-induced vascular resistance, heart rate, and sympathetic activity Alternate splicing of intron 2 generate 4 isoforms of the 5HT 7 receptor (5HT 7A -5HT 7D ) that differ in their C-termini, but neither in their tissue distribution, nor pharmacological and signaling features Gs-Coupled 5HT 7 receptor activates adenylate cyclase AC5 as well as Ca ++ -calmodulin-regulated AC1 and AC8 isoforms. In addition, it can trigger the mitogen-activated protein kinase cascade to activate extracellular signal-regulated protein kinases ERK1 and ERK2 Two active forms of somatostatin derive from alternative cleavage of a single preproprotein: somatostatin 14 202 and somatostatin 28 Agonist stimulation can dissociate P85 regulatory subunit of phosphoinositide 3-kinase (PI3K) and somatostatin receptor Sst 2 , thereby reducing the cell survival PI3K-PKB pathway The HT 4 receptor lodges in basal ganglia, hippocampus, olfactory tubercle, and limbic structures The 5HT 7 receptor is detected in the thalamus, hypothalamus, hippocampus, and cerebral cortex It is produced by neuroendocrine neurons of the periventricular nucleus of the hypothalamus Main transducer Glucagon Gs GLP1, exendins-3/4 Gs GLP2 Gs GIP Gs GHRH Gs Secretin Gs Ghrelin is cleaved from its precursor preproghrelin. Alternative splicing generates a second peptide, des-Gln14-ghrelin with equipotent activity [5] . Both peptides undergo a post-translational octanoylation (Ser3) for full activity on binding to Gq/11-coupled ghrelin receptor (a.k.a. growth hormone secretagog receptor type-1 and growth hormone-releasing peptide receptor) in the hypothalamus and pituitary gland and release of growth hormone from the pituitary gland. Glucagon receptors are activated by glucagon, glucagon-like peptides GLP1 and GLP2, 136 glucose-dependent insulinotropic polypeptide, 137 growth hormonereleasing hormone (GHRH), and secretin.Growth hormone-releasing peptide stimulates growth hormone release via the GH secretagog-receptor GHSR1a, the ghrelin G-protein-coupled receptor. In addition, growth hormone-releasing peptide protects the cardiovascular apparatus independently of growth hormone. Growth hormone releasing-peptide is a ligand for leukocyte differentiation antigen cluster of differentiation CD36, or ScaRb3 scavenger receptor. 138 Transmembrane protein CD36 binds to collagen, thrombospondin, anionic phospholipids, long chain fatty acids, and oxidized LDLs. It is involved in cell adhesion and transport. Growth hormone-releasing peptides enhance the synthesis of ATP-binding cassette transporters ABCa1 and ABCg1 that are responsible for cholesterol efflux from macrophages, hence having a potent anti-atherosclerotic activity (Sect. 6.3.6.4).136. Glucagon as well as GLP1 and GLP2 have a common precursor. 137. A.k.a. gastric inhibitory polypeptide. 138. A.k.a. collagen-1 and thrombospondin receptor, coronary heart disease susceptibility protein CHDS7, fatty acid translocase (FAT), periodic acid-Schiff staining plateletagglutinating substance PAS4, and glycoproteins GP3b, GP4, and GP88.