INTRODUCTION
G protein-coupled receptors (GPCRs) constitute the largest receptor family, orchestrating signaling pathways vital for numerous physiological processes including sensation,1 neural transmission, inflammation, cellular growth and differentiation, muscle contraction and relaxation, and sodium/fluid homeostasis.2 Typically situated on the cell membrane, GPCRs feature seven transmembrane domains (7-TM) structured by three intracellular loops (ILs) and three extracellular loops (ELs), with an intracellular C-terminal and an extracellular N-terminal domain.3 Upon ligand binding, these transmembrane structures undergo conformational changes, facilitating diverse three-dimensional configurations of GPCRs. In addition to single receptor originated downstream signaling, GPCRs can interact with each other, to form dimers or oligomers that synchronize the signaling pathways. While it was initially believed that monomeric GPCRs solely interact with single heterotrimeric G proteins in response to external stimuli, further exploration has revealed a more intricate landscape. Although dimerization is evident in many GPCRs, the specific interactions leading to dimer formation and their significance in pharmacological signaling remain the subjects of ongoing investigation. Notably, a substantial percentage of drugs (~35%) target GPCRs, with over 134 small molecules or peptides approved for use in the USA and the European Union.4 Moreover, a considerable number of GPCRs (~128 out of approximately n 800 known GPCRs) lack known agonists (orphans), rendering them potential candidates for drug discovery efforts, particularly within the context of dimer formation.5 This review aims to elucidate the fundamental aspects of GPCR dimerization within a broader perspective of drug discovery. Additionally, within the renin-angiotensin system, GPCR dimers and oligomers, such as angiotensin receptors, Mas receptors, and bradykinin receptors, play a pivotal role in regulating blood pressure, fluid homeostasis, and the maintenance of electrolyte balance, underscoring their significance in physiological processes. Hence, these receptors have been specifically chosen for detailed examination.
MECHANISMS OF DIMERIZATION
In the typical mode of GPCR signal transduction, upon ligand binding, GPCR interacts with the Gαβγ heterotrimeric protein, facilitating the exchange of GTP with Gα subunit bound GDP. This exchange initiates dissociation of the Gα and Gβγ subunits, which then interact with their respective effector molecules. In humans, 16 genes have been identified to encode 21 Gα subunits that interact with diverse effector molecules.6 For instance, Gαs stimulates adenylate cyclase (AC) activity, leading to increased production of the second messenger cAMP, whereas Gαi inhibits AC, consequently decreasing cAMP levels7 Similarly, Gαq activates phospholipase Cβ (PLCβ), and Gα11 activates Bruton's tyrosine kinase.8 Initially, these interactions were thought to be random, following the “collision coupling model,”9 which aligns with the “fluid mosaic” model of the plasma membrane.10,11 Evidence also suggests the existence of permanent pre-coupled GPCRs and G proteins, as observed in the adenosine 2A receptor. Recent studies have revealed receptor–receptor interactions and cross-talks, indicating their physiological and pharmacological significance in altering signal transduction pathways. This interaction between two identical receptors is termed as homo-dimerization, the interaction between two different receptors is hetero-dimerization and the involvement of more than two receptors is termed oligomerization. For example, Patel et al. (2017) demonstrated that AT2-Mas receptor heterodimer formation in the renin-angiotensin system is facilitated by the close proximity (<7Å) between these two 7-transmembrane receptors with formation of disulfide bridges, particularly under oxidative conditions.12 Some GPCRs, which do not strictly follow G protein coupling, exhibit dimerization or oligomerization involving different transmembrane domains (e.g., TMD VI in β2 adrenoceptor, TMD IV and V in rhodopsin, TMD IV in dopamine) or extracellular domains (e.g., Bradykinin B2 receptor).13–16
Chemically dimerization can be formed by three types of interactions: disulfide bond, coiled-coil interaction, and transmembrane helices interactions (Figure 1), which modulate receptor pharmacology by altering ligand binding affinity and the mode of signal transduction. In classical GPCRs, binding of an allosteric modulator at an allosteric site can change the binding affinity and efficacy of the orthosteric agonist at the same receptor, occurring at two different sites within the same receptor. Homodimerization facilitates the cooperative binding and allosteric modulation of ligands for both receptors (Figure 2). During heteromerization, evidence suggests an increase in receptor activity, such as in the angiotensin II type 1 (AT1)-bradykinin (B2) receptor complex.17 Homodimer formation often leads to the activation of effector molecules without Gαβγ subunit dissociation, while heterodimerization may facilitate coupling to new G proteins, as observed in the δ-Opioid-μ-Opioid heterodimer coupling to Gαz.18–20 Additionally, heterodimerization can result in the coupling of β-arrestin instead of G proteins, initiating the activation of transcription factors by modulating signals.21 Marino et al. (2023) utilized an advanced free-energy technique known as coarse-grained metadynamics to investigate the dimerization process. This method allowed them to simulate the binding events between receptors in a realistic timeframe. They discovered symmetric and asymmetric dimeric structures for CCR5/CCR5, CXCR4/CXCR4, and CCR5/CXCR4 receptors suggesting that these dimerizations work via positive allosteric mechanisms.22
[IMAGE OMITTED. SEE PDF]
[IMAGE OMITTED. SEE PDF]
IMPLICATIONS OF
Signal transduction and modulation
GPCR dimerization, particularly heterodimerization, and oligomerization has the potential to modify G protein coupling and signal transduction pathways. A notable example is adenosine A1 receptor (A1R), a GPCR that regulates neuronal excitability and pain sensitivity. In cerebellar Purkinje cells, A1R forms a trimeric complex with type 1 metabotropic glutamate receptor (mGluR1). Activation of A1R dampens neuronal responses induced by glutamate through mGluR1; mGluR1 activation attenuates A1R signaling, as evidenced by changes in intracellular cAMP levels.23 Another receptor group, the Mas-related receptors (Mrgs), is implicated in pain physiology and serves as a therapeutic target. The heterodimeric interactions of rat MrgD and MrgE alter the MrgD signaling pathway. In human embryonic kidney 293 cell lines, MrgD induces intracellular calcium ion elevation via phosphorylation of the MAP kinases ERK1 and ERK2 in response to beta-alanine, followed by translocation to intracellular vesicles from the plasma membrane. In contrast, MrgE did not respond to beta-alanine. The co-expression of MrgD and MrgE in peripheral nociceptive neurons reveals their heterodimerization, which attenuates MrgD internalization and trafficking, underscoring the significance of dimerization in MrgD function.24 Signal transduction change is also observed in the dimers and oligomers in the renin-angiotensin system (RAS) in angiotensin II receptors (AT1R and AT2R), Mas receptor, relaxin receptors, and adiponectin receptors which is discussed later in this review article.
Hormone resistance
Oligomerization has implications for hormone resistance disorders, such as thyroid stimulating hormone (TSH) resistance in congenital hypothyroidism.25,26 Partial TSH resistance has been observed in cases with heterozygous mutations in the TSH receptor gene.27 However, the underlying molecular mechanisms remain elusive until Calebiro et al. (2005) elucidated the dominant negative effect of mutant TSH receptors on wild-type TSH receptors. This discovery emerged from experiments in which COS-7 cells were co-transfected with wild-type and mutant TSH receptors from TSH-resistant patients. Through FRET and immunoprecipitation studies, oligomerization of these two receptor types in the endoplasmic reticulum was confirmed, shedding light on the basis of TSH resistance.26
Affinity and efficacy of binding
Although pharmacological actions might be reduced in the dimerized state, dimerization can enhance affinity and signaling efficacy through synergism (e.g., μ-δ opioid receptor heterodimers, κ-δ heterodimer, and SSTR5-SSTR1 heterodimer).18,28,29 Heterodimerization also influences receptor trafficking properties, such as surface expression (e.g., GABABR1a and GABABR2), and can lead to either increased (β2-adrenoceptor-δ opioid heterodimer) or decreased (β2-adrenoceptor-κ opioid heterodimer) agonist-mediated endocytosis,28 as well as reduced ligand binding (e.g., A1-D1 heterodimer).30
DRUG DISCOVERY TARGETS
GPCR dimerization is now accepted with some skepticism when a large number of experiments have shown evidence of modulation of receptor functions and the signaling process using the dimerization state- which makes a platform for drug discovery. For example, Adenosine A1 and A2A receptors have opposite signaling pathways that are activated by adenosine binding. These two receptors form hetero dimers in the cortical limbic and thalamic regions of the brain, respectively. At low concentration adenosine inhibits glutamate release by activating A1 receptors, whereas at high concentration facilitates the glutamate release from presynaptic neurons by activating the A2A receptor that has opposite action to its dimer brother A1 receptor.31 This dimeric state can be a tool for the dose-dependent use of A1 receptor agonists to control glutamate release in diseases such as bipolar disorder and depression. Similarly, new compounds are being discovered as dual agonists of heterodimer complexes, for example, Gq/11 coupled D1-D2 receptor complex gets activated by the dual agonist SKF83959, which is a full agonist for D1 and a partial agonist for D2 receptors.32 This simultaneous activation is mandatory for G-protein coupling and intracellular calcium release by this oligomeric complex. Likewise, Vendrell et al. (2007) developed an ergopeptide derivative capable of acting as an agonist for the adenosine receptor and antagonist of the dopamine receptor,33 which can be used to investigate adenosine-dopamine heterodimers. A TMD4 peptide-inhibiting dimer in the chemokine receptor CXCR4 has shown potential for targeting some inflammatory conditions.34 With advances in pharmacology, biased pathways (e.g., β arrestin) can be activated using biased ligands. Gαq, Gαi2, Gα12, and Gα13 activities can be reduced by stimulating Orexin-1/Cholecystokinin A receptor heterodimers using their double ligands.35 Thus it would not be an exaggeration to say that, when clinical studies are being performed on a new drug candidate, the consideration of receptor dimerization might open new targets and increase the specificity of drugs at the molecular level. In addition, state-of-the-art techniques such as co-immunoprecipitation, Western blotting, bioluminescence or fluorescence resonance energy transfer (BRET or FRET), receptor labeling, microscopy, proximity ligation assay (PLA), and protein fragment complementation assay (PCA) are used to identify homo and heterodimers and drug discovery targets.36–42
TARGETING DIMERIZATION IN RENIN-ANGIOTENSIN SYSTEM (
Angiotensin II Type I (AT1) receptors, found in the kidney, heart, liver, lung, brain, and vascular system,43 are implicated in kidney injury, blood pressure control, and fluid homeostasis, and can form dimers, initially which was thought to be protein aggregates (Milligan and Bouvier, 2005; Siemens et al., 1991; Rondeau et al., 1990). The BRET study and regulated secretion and aggregation assay (RPD)44 showed that AT1 receptors form constitutive homo-dimers and oligomers in COS-7 cells. This BRET signal was independent of the agonist angiotensin II agonist and the inverse agonist telmisartan. Simultaneously, the regulated secretion and aggregation technology enabled them to regulate the escape of AT1 receptors to the cell surface from the endoplasmic reticulum (ER) by fluorescent Fm protein tagging to introduce AT1-Fm aggregation. Treatment with the aggregation attenuator drug AP21998 revealed that surface expression of AT1 receptors can be controlled with this drug if AT1 is co expressed with mutated (in DRY motif) AT1 receptor tagged to the Fm protein.45 This study also revealed that wild-type AT1, and mutant AT1 receptor dimerization lose the capacity to couple Gαq but can preserve ERK activation, indicating the importance of homodimerization in AT1 receptor activity. Similarly, AT1 homodimerization in monocytes has been used as a diagnostic tool for atherogenesis. The intracellular factor XIIIA transglutaminase creates cross-linking with the carboxy terminal end of the AT1 homodimer via glutamine in the presence of angiotensin II, showing desensitization and modulation of receptor signaling.46 Inhibition of ACE and XIIIA transglutaminase showed less cross-linked AT1 receptors in an ApoE deficient mouse atherosclerosis model, which opens a window for screening drug molecules for treating arterial diseases.47–49 Thus, targeting AT1 homodimers could be a therapeutic intervention for various disease conditions.
Angiotensin II type II (AT2) receptors are expressed in the tissues of various organs such as the fetus, kidney, heart, lung, adrenal cortex, uterus, and vascular smooth muscle.43 AT2 receptors are involved in reducing hypertension, metabolic dysfunction, inflammation, and organ remodeling.50,51 They reduce tubular damage by reducing the infiltration of IL-6, TNF-α, and CD11b+ leukocytes in acute kidney injury in renal epithelial cells.52 They also decrease PKC activation and superoxide formation in neuronal cells and enhance NO availability and cerebral vasorelaxation in ischemic stroke.53,54 AT2 receptors can form constitutive dimers with two cysteines (at the 35N terminus and 290 position in ECL3), which are crucial for dimerization.55 Mutations in these positions can halt signaling, which might affect the protective actions of AT2 receptors in the body.56,57
Vasoactive AT1 and Bradykinin (B2) receptor heterodimers have been found ubiquitously in number of several cell lines, platelets, neurons, and vascular smooth muscle.17,58 While the AT1 receptor is related to hypertension and cardiovascular pathophysiology, the AT1/B2 dimer showed hyper-responsiveness in the AT1 agonist Ang II- mediated coupling in Gαq/11 and elevated secretion of endothelin-1 from mesangial cells in a hypertensive rat model.58 This dimer has also been found in hypertensive preeclamptic patients in a clinical settings of 30 pregnant women including 19 preeclampsia patients.59 This AT1/B2 dimer state correlates with Ang II hypersensitivity and a 5-fold increase in B2 protein expression, which makes this AT1/B2 dimers a selective target for new drug candidates in hypertension and pre-eclampsia.
Abadir et al. (2006) found the presence of a constitutive AT2/B2 heterodimer in PC12W cells showing dimer entanglement in the NO production mechanism. This heterodimer also contributes to the phosphorylation of c-Jun N-terminal kinase, κBα, phosphotyrosine phosphatase, and dephosphorylation of P42/44 mitogen activated protein kinase and P38.60 As these proteins are well known for different physiological processes, they provide a chance for the therapeutic intervention of AT2 and B2 receptor expression in cardiovascular and renal diseases.
AT1 and AT2 receptors show opposite effects in the renin-angiotensin system by antagonizing each other's signal transduction.61 The AT1/AT2 heterodimer is functionally relevant to the signaling pattern. Ang II forms complex with the AT1/AT2 heterodimer in the LLC-PK1 cell membrane and this complex is internalized through the microtubule-dependent endocytic pathway and localizes in the ER. This process also stimulates the intracellular Ca+2 response by elevating sarcoplasmic reticulum Ca+2-ATPase activity.62 Hence, the ER can be a target organelle for RAS (Table 1).
TABLE 1 Dimers in renin-angiotensin system, and their characteristics.
| Name of dimers/oligomer | Characteristics | Condition when dimer/oligomer might form | Organ/Cells/Animal model |
| AT1R/AT1R | AT1 receptor dimerization loses the capacity to couple Gαq but can preserve ERK activation45 | Normal, Atherogenesis48 | Human kidney, heart, liver, lung, brain, and vascular system43,47 |
| AT2R/AT2R | Enhances AT2R mediated signaling and protection55 | Normal condition | Human fetus, kidney, heart, lung, adrenal cortex, uterus, and vascular smooth muscle43 |
| AT1R/B2R | Increases AngII hypersensitivity in Gαq/11 coupling58 | Hypertension,58 pre-eclampsia59 | Human vascular smooth muscle, platelet, neurons17,58 |
| AT2R/B2R | Increases NO production, phosphorylation of JNKs, κBα, phosphotyrosine phosphatase, and dephosphorylation of P42/44 MAPK and P3860 | PC12W cell line60 | |
| AT1R/AT2R | Stimulates the intracellular Ca+2 response by elevating sarcoplasmic reticulum Ca+2-ATPase activity62 | Normal condition especially in the presence of Ang II | LLC-PK1 cell62 |
| AT2R/MasR | Enhances receptor functions, shows functional interdependence and crosstalks12 | Obesity | Obese Zucker rat kidney12 |
| AT2R/RXFP-1 | Regulates the anti-fibrotic effect of relaxin hormone after the dimer is formed63 | In fibrotic conditions like CKD | Mouse model of unilateral ureteral obstruction63 |
| AT1R/AT2R/RXFP-1 | AT1R regulates the anti-fibrotic actions of AT2R or RXFP-164 | Renal and cardiac fibrosis | Cardiac and renal myofibroblasts64 |
| AT1R/Adipo R1 and AT2R/Adipo R2 | Induce tubulointerstitial fibrosis65 | Diabetic kidney disease, high glucose | Renal proximal tubular epithelial cells (NRK-52E)65 |
AT2 and Mas receptors showed functional cross-talks and physical heterodimerization in obese Zucker rat kidneys, which was confirmed by co-IP, antibody labeling, confocal microscopy, and immunoblotting.12,66 Patel et al. (2017) showed that the AT2R agonist drug C21 promoted natriuretic function in an obese Zucker rat model, as confirmed by increased sodium and urine flow which was lowered by concomitant administration of the AT2R antagonist PD123319 or MasR antagonist A-779 drugs. Similar results were observed with the infusion of MasR agonist Ang-(1-7) and prior infusion of PD123319 or A-779. This heterodimerization, cross talks, and reciprocal modulation of AT2 and MasR have made them potential targets for new drug molecules such as C21 and it's possible derivatives.
Relaxin hormone reduces fibrosis mediated by transforming growth factor-β1 (TGF-β1) by repealing Smad-2 phosphorylation via relaxin family peptide receptor −1 (RXFP-1). Angiotensin II also reduces the TGF- β1 activity via AT2R. Previous studies showed anti-fibrotic action by AT2R activation in the kidney, lung, heart, skin, and pancreas.67 Interestingly, heterodimerization of AT2R/RXFP-1 has been identified using both animal and in-vitro models.63 The AT2R blocker PD123319 blocked the anti-fibrotic effect of relaxin in a mice model of unilateral ureteral obstruction,63 indicating the importance of AT2R/RXFP-1 dimer in the anti-fibrotic action of relaxin in chronic kidney disease (CKD). A recent study revealed that relaxin also inhibits NLRP3 inflammasome in cardiac myofibroblast via AT2R and ATP receptors (P2X7).68 The existence of AT1R/AT2R/RXFP-1 oligomer in cardiac and renal myofibroblasts and the loss of AT2R or RXFP-1 mediated anti-fibrotic actions by AT1R blocker irbesartan and candesartan suggested therapeutic interventions targeting these oligomers.64
Adiponectin receptors (Adipo R1 and Adipo R2) show higher expression in diabetic kidney disease by forming heterodimers with the AngII receptors: AT1/Adipo R1 and AT2/Adipo R2 in renal proximal tubular epithelial cells (NRK-52E) in response to high glucose.65 These heterodimers induced tubulointerstitial injury in kidney. Hence, targeting these heterodimers might have therapeutic potential in kidney diseases.
CONCLUDING REMARKS
GPCR dimerization or oligomerization changes signaling processes, which creates a big question: should we not reconsider receptor pharmacology in drug discovery? Although the dimerization concept has not yet gained interest in the pharmaceutical industry, there is enormous potential for finding new dimers or oligomeric targets for treating life-threatening diseases such as hypertension or atherosclerosis. Targeting biased signaling from GPCR dimers might give more information about disease conditions, cross-talks among GPCRs, and any possible side effects. Hence, GPCR dimerization studies can be worthy of consideration in drug discovery.
AUTHOR CONTRIBUTIONS
TF: conceptualization, writing, review, and editing the manuscript, editing; TH: conceptualization, review, and editing the manuscript, funding acquisition.
ACKNOWLEDGMENTS
This work is supported by National Institutes of Health R01 grants DK061578 and DK117495.
CONFLICT OF INTEREST STATEMENT
The authors declare that there is no potential financial or commercial conflict of interest with this article.
DATA AVAILABILITY STATEMENT
The data that support the findings of this study are available from the studies mentioned in the references, and the authors can also provide any data upon request.
Adler E, Hoon MA, Mueller KL, Chandrashekar J, Ryba NJ, Zuker CS. A novel family of mammalian taste receptors. Cell. 2000;100:693‐702.
Calebiro D, Godbole A. Internalization of G‐protein‐coupled receptors: implication in receptor function, physiology and diseases. Best Pract Res Clin Endocrinol Metab. 2018;32:83‐91.
Bouvier M. Oligomerization of G‐protein‐coupled transmitter receptors. Nat Rev Neurosci. 2001;2:274‐286.
Sriram K, Insel PA. G protein‐coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol. 2018;93:251‐258.
Alexander SPH, Christopoulos A, Davenport AP, et al. THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein‐coupled receptors. Br J Pharmacol. 2019;176(Suppl 1):S21‐S141.
Oldham WM, Hamm HE. Heterotrimeric G protein activation by G‐protein‐coupled receptors. Nat Rev Mol Cell Biol. 2008;9:60‐71.
Jiang H, Galtes D, Wang J, Rockman HA. G protein‐coupled receptor signaling: transducers and effectors. Am J Physiol Cell Physiol. 2022;323:C731‐C748.
Cabrera‐Vera TM, Vanhauwe J, Thomas TO, et al. Insights into G protein structure, function, and regulation. Endocr Rev. 2003;24:765‐781.
Tolkovsky AM, Levitzki A. Mode of coupling between the beta‐adrenergic receptor and adenylate cyclase in Turkey erythrocytes. Biochemistry. 1978;17:3795‐3810.
Singer SJ, Nicolson GL. The fluid mosaic model of the structure of cell membranes. Science. 1972;175:720‐731.
Ferré S. The GPCR heterotetramer: challenging classical pharmacology. Trends Pharmacol Sci. 2015;36:145‐152.
Patel SN, Ali Q, Samuel P, Steckelings UM, Hussain T. Angiotensin II type 2 receptor and receptor mas are colocalized and functionally interdependent in obese Zucker rat kidney. Hypertension. 2017;70:831‐838.
Hebert TE, Moffett S, Morello JP, et al. A peptide derived from a beta2‐adrenergic receptor transmembrane domain inhibits both receptor dimerization and activation. J Biol Chem. 1996;271:16384‐16392.
Fotiadis D, Jastrzebska B, Philippsen A, Müller DJ, Palczewski K, Engel A. Structure of the rhodopsin dimer: a working model for G‐protein‐coupled receptors. Curr Opin Struct Biol. 2006;16:252‐259.
Lee SP, O'Dowd BF, Rajaram RD, Nguyen T, George SR. D2 dopamine receptor homodimerization is mediated by multiple sites of interaction, including an intermolecular interaction involving transmembrane domain 4. Biochemistry. 2003;42:11023‐11031.
Michineau S, Alhenc‐Gelas F, Rajerison RM. Human bradykinin B2 receptor sialylation and N‐glycosylation participate with disulfide bonding in surface receptor dimerization. Biochemistry. 2006;45:2699‐2707.
AbdAlla S, Lother H, Quitterer U. AT1‐receptor heterodimers show enhanced G‐protein activation and altered receptor sequestration. Nature. 2000;407:94‐98.
Gomes I, Jordan BA, Gupta A, Trapaidze N, Nagy V, Devi LA. Heterodimerization of mu and delta opioid receptors: a role in opiate synergy. J Neurosci. 2000;20:Rc110.
Jordan BA, Cvejic S, Devi LA. Opioids and their complicated receptor complexes. Neuropsychopharmacology. 2000;23:S5‐S18.
Fan T, Varghese G, Nguyen T, Tse R, O'Dowd BF, George SR. A role for the distal carboxyl tails in generating the novel pharmacology and G protein activation profile of mu and delta opioid receptor hetero‐oligomers. J Biol Chem. 2005;280:38478‐38488.
Rozenfeld R, Devi LA. Receptor heteromerization and drug discovery. Trends Pharmacol Sci. 2010;31:124‐130.
Di Marino D, Conflitti P, Motta S, Limongelli V. Structural basis of dimerization of chemokine receptors CCR5 and CXCR4. Nat Commun. 2023;14:6439.
Kamikubo Y, Tabata T, Sakairi H, Hashimoto Y, Sakurai T. Complex formation and functional interaction between adenosine A1 receptor and type‐1 metabotropic glutamate receptor. J Pharmacol Sci. 2015;128:125‐130.
Milasta S, Pediani J, Appelbe S, et al. Interactions between the mas‐related receptors MrgD and MrgE alter signalling and trafficking of MrgD. Mol Pharmacol. 2006;69:479‐491.
Dumitrescu AM, Korwutthikulrangsri M, Refetof S. In: Feingold KR, Anawalt B, Blackman MR, et al., eds. Impaired Sensitivity to Thyroid Hormone: Defects of Transport, Metabolism, and Action. Endotext; 2000.
Calebiro D, de Filippis T, Lucchi S, et al. Intracellular entrapment of wild‐type TSH receptor by oligomerization with mutants linked to dominant TSH resistance. Hum Mol Genet. 2005;14:2991‐3002.
Huang B, Liu C, Xiao F, et al. Resistance to thyroid hormone caused by heterozygous mutation of thyroid hormone receptor B gene c.G1378A: report of one Chinese pedigree and literature review. Clin Case Rep. 2021;9:1055‐1059.
Jordan BA, Devi LA. G‐protein‐coupled receptor heterodimerization modulates receptor function. Nature. 1999;399:697‐700.
Rocheville M, Lange DC, Kumar U, Patel SC, Patel RC, Patel YC. Receptors for dopamine and somatostatin: formation of hetero‐oligomers with enhanced functional activity. Science. 2000;288:154‐157.
Ginés S, Hillion J, Torvinen M, et al. Dopamine D1 and adenosine A1 receptors form functionally interacting heteromeric complexes. Proc Natl Acad Sci USA. 2000;97:8606‐8611.
Ciruela F, Ferré S, Casadó V, et al. Heterodimeric adenosine receptors: a device to regulate neurotransmitter release. Cell Mol Life Sci. 2006;63:2427‐2431.
Rashid AJ, So CH, Kong MM, et al. D1‐D2 dopamine receptor heterooligomers with unique pharmacology are coupled to rapid activation of Gq/11 in the striatum. Proc Natl Acad Sci USA. 2007;104:654‐659.
Vendrell M, Angulo E, Casadó V, et al. Novel ergopeptides as dual ligands for adenosine and dopamine receptors. J Med Chem. 2007;50:3062‐3069.
Wang J, Norcross M. Dimerization of chemokine receptors in living cells: key to receptor function and novel targets for therapy. Drug Discov Today. 2008;13:625‐632.
Bai B, Chen X, Zhang R, et al. Dual‐agonist occupancy of orexin receptor 1 and cholecystokinin a receptor heterodimers decreases G‐protein‐dependent signaling and migration in the human colon cancer cell line HT‐29. Biochim Biophys Acta, Mol Cell Res. 2017;1864:1153‐1164.
Suzuki T, Namba K, Yamagishi R, Kaneko H, Haga T, Nakata H. A highly conserved tryptophan residue in the fourth transmembrane domain of the a adenosine receptor is essential for ligand binding but not receptor homodimerization. J Neurochem. 2009;110:1352‐1362.
Popa MO, Lerche H. Cu2+ (1,10 phenanthroline)3 is an open‐channel blocker of the human skeletal muscle sodium channel. Br J Pharmacol. 2006;147:808‐814.
Alam MS. Proximity ligation assay (PLA). Curr Protoc Immunol. 2018;123: [eLocator: e58].
López‐Cano M, Fernández‐Dueñas V, Ciruela F. Proximity ligation assay image analysis protocol: addressing receptor‐receptor interactions. Methods Mol Biol. 2019;2040:41‐50.
Michnick SW, Remy I, Campbell‐Valois FX, Vallée‐Bélisle A, Pelletier JN. Detection of protein‐protein interactions by protein fragment complementation strategies. Methods Enzymol. 2000;328:208‐230.
Bardiya N, Alexander WG, Perdue TD, et al. Characterization of interactions between and among components of the meiotic silencing by unpaired DNA machinery in Neurospora crassa using bimolecular fluorescence complementation. Genetics. 2008;178:593‐596.
Zamyatnin AA Jr, Solovyev AG, Bozhkov PV, Valkonen JP, Morozov SY, Savenkov EI. Assessment of the integral membrane protein topology in living cells. Plant J. 2006;46:145‐154.
Capponi AM. Distribution and signal transduction of angiotensin II AT1 and AT2 receptors. Blood Press Suppl. 1996;2:41‐46.
Rivera VM, Wang X, Wardwell S, et al. Regulation of protein secretion through controlled aggregation in the endoplasmic reticulum. Science. 2000;287:826‐830.
Hansen JL, Theilade J, Haunsø S, Sheikh SP. Oligomerization of wild type and nonfunctional mutant angiotensin II type I receptors inhibits galphaq protein signaling but not ERK activation. J Biol Chem. 2004;279:24108‐24115.
AbdAlla S, Lother H, Langer A, el Faramawy Y, Quitterer U. Factor XIIIA transglutaminase crosslinks AT1 receptor dimers of monocytes at the onset of atherosclerosis. Cell. 2004;119:343‐354.
Al‐Horani RA, Kar S. Factor XIIIa inhibitors as potential novel drugs for venous thromboembolism. Eur J Med Chem. 2020;200: [eLocator: 112442].
Yamada M, Kushibiki M, Osanai T, Tomita H, Okumura K. Vasoconstrictor effect of aldosterone via angiotensin II type 1 (AT1) receptor: possible role of AT1 receptor dimerization. Cardiovasc Res. 2008;79:169‐178.
Lyngsø C, Erikstrup N, Hansen JL. Functional interactions between 7TM receptors in the renin‐angiotensin system—dimerization or crosstalk? Mol Cell Endocrinol. 2009;302:203‐212.
Fatima N, Patel SN, Hussain T. Angiotensin II type 2 receptor: a target for protection against hypertension, metabolic dysfunction, and organ remodeling. Hypertension. 2021;77:1845‐1856.
Fatima N, Ali R, Faisal T, Kulkarni K, Patel S, Hussain T. Macrophage angiotensin AT(2) receptor activation is protective against early phases of LPS‐induced acute kidney injury. Am J Physiol Renal Physiol. 2023;325:F552‐f563.
Matavelli LC, Huang J, Siragy HM. Angiotensin AT₂ receptor stimulation inhibits early renal inflammation in renovascular hypertension. Hypertension. 2011;57:308‐313.
McCarthy CA, Vinh A, Callaway JK, Widdop RE. Angiotensin AT2 receptor stimulation causes neuroprotection in a conscious rat model of stroke. Stroke. 2009;40:1482‐1489.
Min LJ, Mogi M, Tsukuda K, et al. Direct stimulation of angiotensin II type 2 receptor initiated after stroke ameliorates ischemic brain damage. Am J Hypertens. 2014;27:1036‐1044.
Miura S, Karnik SS, Saku K. Constitutively active homo‐oligomeric angiotensin II type 2 receptor induces cell signaling independent of receptor conformation and ligand stimulation. J Biol Chem. 2005;280:18237‐18244.
Miura S, Zhang J, Boros J, Karnik SS. TM2‐TM7 interaction in coupling movement of transmembrane helices to activation of the angiotensin II type‐1 receptor. J Biol Chem. 2003;278:3720‐3725.
Steckelings UM, Widdop RE, Sturrock ED, et al. The angiotensin AT(2) receptor: from a binding site to a novel therapeutic target. Pharmacol Rev. 2022;74:1051‐1135.
AbdAlla S, Abdel‐Baset A, Lother H, el Massiery A, Quitterer U. Mesangial AT1/B2 receptor heterodimers contribute to angiotensin II hyperresponsiveness in experimental hypertension. J Mol Neurosci. 2005;26:185‐192.
AbdAlla S, Lother H, el Massiery A, Quitterer U. Increased AT(1) receptor heterodimers in preeclampsia mediate enhanced angiotensin II responsiveness. Nat Med. 2001;7:1003‐1009.
Abadir PM, Periasamy A, Carey RM, Siragy HM. Angiotensin II type 2 receptor‐bradykinin B2 receptor functional heterodimerization. Hypertension. 2006;48:316‐322.
Miura S, Matsuo Y, Kiya Y, Karnik SS, Saku K. Molecular mechanisms of the antagonistic action between AT1 and AT2 receptors. Biochem Biophys Res Commun. 2010;391:85‐90.
Ferrão FM, Cardoso LHD, Drummond HA, et al. Luminal ANG II is internalized as a complex with AT(1)R/AT(2)R heterodimers to target endoplasmic reticulum in LLC‐PK(1) cells. Am J Physiol Renal Physiol. 2017;313:F440‐f449.
Chow BS, Kocan M, Bosnyak S, et al. Relaxin requires the angiotensin II type 2 receptor to abrogate renal interstitial fibrosis. Kidney Int. 2014;86:75‐85.
Chow BSM, Kocan M, Shen M, et al. AT1R‐AT2R‐RXFP1 functional crosstalk in myofibroblasts: impact on the therapeutic targeting of renal and cardiac fibrosis. J Am Soc Nephrol. 2019;30:2191‐2207.
Zha D, Cheng H, Li W, et al. High glucose instigates tubulointerstitial injury by stimulating hetero‐dimerization of adiponectin and angiotensin II receptors. Biochem Biophys Res Commun. 2017;493:840‐846.
Leonhardt J, Villela DC, Teichmann A, et al. Evidence for heterodimerization and functional interaction of the angiotensin type 2 receptor and the receptor MAS. Hypertension. 2017;69:1128‐1135.
Sumners C, Peluso AA, Haugaard AH, Bertelsen JB, Steckelings UM. Anti‐fibrotic mechanisms of angiotensin AT(2) ‐receptor stimulation. Acta Physiol (Oxf). 2019;227: [eLocator: e13280].
Tapia Cáceres F, Gaspari TA, Hossain MA, Samuel CS. Relaxin inhibits the cardiac myofibroblast NLRP3 inflammasome as part of its anti‐fibrotic actions via the angiotensin type 2 and ATP (P2X7) receptors. Int J Mol Sci. 2022;23: [eLocator: 7074].
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025. This work is published under http://creativecommons.org/licenses/by-nc/4.0/ (the "License"). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
G protein‐coupled receptors (GPCRs), which play crucial roles in various physiological functions, often assembled into dimers and higher‐order oligomers. This oligomerization phenomenon has been observed in diverse physiological and pathological contexts, presenting promising opportunities for drug discovery targeting vital systems such as the cardiovascular, nervous, endocrine, and renal systems. This review offers a concise understanding of GPCR dimerization, its signaling mechanisms, and its implications. Furthermore, we explored therapeutic strategies aimed at modulating receptors involved in dimer/oligomer formation within the renin‐angiotensin system.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer





